DTMH Flashcards

1
Q

Global burden of dengue

A

390 million worldwide per year
40-50% of world at risk
70% cases in Asia
Longer and more wet rainy seasons
Cases in Americas rising

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Virus of dengue

A

RNA virus
4 serotypes
3 structural proteins (C, M, E)
7 non-structural proteins (NS)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Process of dengue replication

A

Binds to receptors
Clarity in mediated endocytosis
Fusion and virus assembly
Viral RNA released into cell
Translation
Viral assembly
Virus maturation
DENV release from cell

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Vectors of dengue

A

Aèdes aegypti:
violin/lyre shaped white markings on thorax
Can bite multiple times

Aedes albopictus
Single silvery white line down thorax
Very adaptable

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Clinical manifestations of dengue

A

incubation 5-7 days
Fever, headache, rashes, muscle pain and joint pain

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Rashes of dengue

A

Initially maculopapular rash
Then petichael rash often at extremities ie ankles
Recovery rash: blanching

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Disease phases dengue

A

febrile phase
Critical period
Recovery phase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Warning signs of dengue

A

Abdominal pain or tenderness
Persistent vomiting
Clinical fluid accumulation
Mucosal bleed
Lethargy or restlessness
Liver enlargement (2+cm)
Increase in Hct concurrent with drop in Plt count

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Definition of acute rheumatic fever

A

acute multi system, inflammatory process occurring 2-3 weeks after grpA streptococcal pharyngitis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Definition of rheumatic heart disease

A

chronic process due to scarring of valves during ARF associated with complications such as HF and death

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Properties of group A streptococcus

A

Streptococcus pyogènes, gram positive, beta-haemolytic cocci in chains

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Clinical manifestations of streptococcus pyogenes

A

Superficial: pharyngitis and pyoderma/impetigo

Invasive/toxin mediated:
Skin, soft tissue, lung
Toxic shock syndrome
Necrotising fasciitis
Puerperal sepsis
Scarlet fever

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Post infective disease manifestations of group A strep

A

Rheumatic fever
Syndenham’s chorea
RHD
Acute glomerulonephritis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Global burden of group A strep

A

33mill worldwide prevalence estimates from echo screening and/or from HF/stroke/arrhythmias/pregnancy related complication data

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Clinical features of group A strep infection

A

Children aged 5-15 years
Joint pain (migratory, large joint)
Carditis (mitral first)
Chorea
Incubation: 10-14days following GAS pharyngitis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Drivers of rheumatic heart disease

A

Group a streptococcus is the trigger but
Kids affected - lack of immunity
Pathogenesis poorly understood
Issues with molecular mimicry - affects vaccine development
Frequency of GrpA strep infections affecting moving on to rheumatic fever - ie poverty/environments can mean some kids are exposed to over 5 different types which then drives up risk, whereas in UK may be much less

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Criteria for RHD
Low risk countries

A

JONES 2015 in low risk
Major:
Carditis
Arthritis - poly
Chorea
Subcutaneous nodules
Erythema marginatum

Minor
Polyarthralgia
Fever 38.5+
ESR 60+/CRP 30+
Prolonged PR

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Criteria for RHD
High risk countries

A

JONES 2015 in high risk
Major:
Carditis
Arthritis - poly or mono, polyarthralgia
Chorea
Subcutaneous nodules
Erythema marginatum

Minor
Monoarthralgia
Fever 38.0+
ESR 60+/CRP 30+
Prolonged PR

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

How to diagnose Rheumatic fever?

A

Evidence of Strep A plus
2x major criteria
1x major and 2x minor

From Jones criteria 2015

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

How to confirm Rheumatic fever?

A
  1. Evidence of Strep A:
    a. Swab, serology, rapid test (except for chorea & indolent carditis)
    1. Serologic tests
      a. ASO & ADB - rising titre
      b. Variable availability and normal ranges
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Diagnosis?
- 7year old girl presents to health centre with SOB + left knee pain + abdominal pain, biventricular failure on CXR, tender gross hepatomegaly, died of cardiogenic shock

A

RHD

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Spooning / milkmaids grip a clinical feature of…

A

Chorea associated with RHD

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Management of rheumatic fever

A

Acute mx:
1. Patient/family education
2. Single dose BenPen or otherwise oral pen or erythromycin if pen allergic
3. Analgesia +/- NSAIDs
4. Diuresis, ACEi, +/- steroids

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Longer term management of rheumatic heart disease

A

Longer term:
1. Prevent recurrence (including sub clinical)
○ Secondary prophylaxis for prevention of ARF = secondary prevention
2. Manage HF
3. Anti coagulation for AF or valve replacement
4. Assessment for intervention/surgery

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Critical diagnostic test for RHD

A

Echocardiogram

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What does secondary prevention mean in the context of RHD?

A

Secondary prophylaxis for prevention of ARF

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

What is management approach to latent RHD?

A

Secondary Abx prophylaxis with penicillin
Trial: GOAL, NEJM
No prophylaxis vs IM Pen 4 weekly
Pen group had 10 times less echo progression of latent disease at 2 years

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

What is the evidence base for anticoagulation in RHD?

A

INVICTUS study, RCT
NEJM
Rivaroxaban was associated with more stroke, emboli, MI, death compared to Vit K antagonist eg warfarin
(24 countries)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Where is yellow fever found? What vector?

A

South America and Africa
Aedes mosquito

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

Describe protozoa

A
  • Taxonomic group of single-celled organisms with a nucleus (ie. Eukaryotes) that are non-photosynthetic
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

Define protista

A

Taxonomic group of single-celled eukaryotes including Protozoa, algae and slime moulds

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

Transmission of protozoa

A

Direct
* Passed directly from one infected host to another by some physical means eg faecal contamination of food or water

Via intermediate host or vector
* Very common amongst parasites
* Sometimes just mechanical transfer, but usually one or more essential life-cycle stages take place here

Definitive host: stage where the sexual stages of the life cycle take place
* May or may not be the human

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

Transmission of protozoa

A
  • Direct
    ○ Passed directly from one infected host to another by some physical means eg faecal contamination of food or water
    • Via intermediate host or vector
      ○ Very common amongst parasites
      ○ Sometimes just mechanical transfer, but usually one or more essential life-cycle stages take place here
      ○ Definitive host: stage where the sexual stages of the life cycle take place
      § May or may not be the human
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

Define anthroponosis and give examples

A

Disease spread from humans to humans
Eg smallpox, malaria

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

Define zoonosis and give examples

A

Any disease which can be transmitted to humans from animals
- E.g. East African sleeping sickness
- These animals form reservoir hosts, which can greatly complicate control or elimination

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

Define trophozoite

A
  • a general, rather ill-defined term for the feeding, growing and dividing stage of a protozoan
    • Note that many organisms have specific names for life-cycle stages that could be described as trophozoites
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

Define cyst in parasitology

A
  • A microscopic, resistant form shed into the environment and responsible for transmission (e.g. Entamoeba)

In the tissue, a distinct walled off cavity containing multiple organisms, often important in transmission (e.g. Trichinella, Toxoplasma). Related to general clinical usage.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

Examples of protozoa that are dangerous for immunocompromised host

A

Toxoplasma gondii
Cryptosporidium
Acanthamoeba species
Balamuthia madrillaris

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

Examples of fungi dangerous to immunocompromised host

A

Pneumocystis jirovecii
Encephalitozoon intestinalis - microsporidiosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

Examples of gut protozoa

A

Giardia intestinalis
Entamoeba histolytica
Babesia microti

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

Newer definitions for symbiosis type relationships

A
  • Mutualism: both benefit
  • Neutralism: neither incur cost or benefit
  • Commensalism: one benefits but at no cost to the other
  • Parasitism: one benefits at the cost of the other
    ○ All pathogens now fit in here
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

Issues with whether something is a pathogen or not

A

Host immunocompetence
Symbionts
Ectopic infection
Presence of other organisms
Microbiome
Coinfections
?theory of dysbiosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

Two relevant phylums of parasitic helminths in medical parasitology

A

Platyhelminthes (flatworms)

Nematoda (round worms)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

Two relevant classes of Phylum Platyhelminthes

A

Trematoda
- flukes

Cestoida
- tapeworms

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

Second biggest cause of human parasitic diseases

A

Schistosomiasis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

Define metazoan and give example

A

Multicellular eukaryotic animal eg parasitic helminths

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

Characteristics of parasitic helminths

A

Complex metazoan
Most multicellular but some can be intracellular
Many are zoonosis -> control is more difficult

  • Infection is by:
    ○ Ingestion of eggs/cysts on/in food (e.g vegetables, meat, fish etc)
    ○ Penetration of skin by infective larval stages (directly or via biting insect)
  • Larvae grow, moult, mature to adult and then produce offspring (eggs or larvae) which are voided from the host to infect new hosts
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

What does a direct helminth life cycle mean?

A

Parasite requires one host to complete its life cycle

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

What does an indirect helminth life cycle mean?

A

parasite requires TWO or MORE HOSTs to complete its life cycle

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What is a definitive host in parasitic life cycle?

A

Host harbouring the sexually mature adult worm

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

What is an intermediate host in parasitic life cycle?

A

Host harbouring the larval stages

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

What determines worm burden that human experiences in helminths infections and what does this determine?

A

Level and duration of exposure ie cumulation

Worm burden determines clinical disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

Examples of diseases caused by helminth infections

A

Schistosomiasis - hepatosplenomegaly + peri-portal fibrosis
Hydatid cyst
Filariasis - elephantiasis
Onchocerciasis
Hookworm -anaemia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

Trematode characteristics

A

Leaf-shaped flatworms - no body cavity, oral and ventral suckers for attachment and sucking nutrients, blind sac-like gut
‘Flukes’
Found in various tissues
All belong to sub-class Digenea ‘digenetic trematodes’
○ Means 2 or more generations in different hosts to complete the life cycle

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

Most important human infecting trematodes

A

Schistosomes (blood fluke)
• Paragonimus (lung fluke)
• Opistorchis/Clonorchis (liver fluke)
• Fasciola (liver fluke)
• Fasciolopsis (intestinal fluke)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

First intermediate host for all trematode schistosome infections

A

Freshwater snail

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

Most important human trematode?

A

Schistosomes
- Morphologically adapted to live inside blood vessels and dioecious (separate sexes)
- Below = Schistosoma spp. = blood fluke
○ Causes Schistosomiasis
○ Affects ~250mill people worldwide
○ Eggs become lodged in tissues -> immune reaction generate granulomas and fibrosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

Common name for Cestoidea

A

Tapeworms

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

Most important human-infecting cestodes

A

Taenia saginata (beef tapeworm)
• Taenia solium (pork tapeworm) (larval infection=cysticercosis)
• Dihyllobothrium spp (fish tapeworm)
• Echinococcus spp (dog and fox tapeworms) (larval infection=hydatid cyst)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

Characteristics of cestodes

A

Segmented ribbon-like (tape) flatworms
- All species of cestodes are parasitic
- Adult worms live in the small intestine of vertebrate animals (definitive host) Larval stages live in tissues of vertebrates and invertebrates (intermediate host)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

What type of helminth has a full intestinal system?

A

Nematodes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

What disease does taenia solium cause?

A

Cysticercosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

Most important human-infecting nematodes:

A

Intestinal nematodes:
• Hookworms
• Ascaris
• Trichuris
• Strongyloides

Tissue nematodes:
• Wuchereria/Brugia (Lymphatic filariasis)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

Basic characteristics of falciparum

A

Most dangerous + most deaths
Pre-erythrocytic stage: 5.5-7 days
Incubation period of 9-14 days

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

Which strains of malaria can be dormant as hyponozoites?

A

ovale and vivax

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

Rare zoonosis malaria strain? Where is it found? Whats the natural host?

A

Knowlesi
Malaysia
Macaques natural host
Pre-erythrocytic 8-9 days
Erythrocytic: 24 hr cycle
Incubation period: 9-12 days

Fastest growth of all human infecting plasmodium

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

Hallmark symptom of malaria and what do changes in it correspond to?

A

Fever
Spikes correspond to erythrocyte cycle and syncronisation of developmental stages

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

What does the threshold at which symptoms develop in malaria depend on?

A

Endemicity, transmission intensity and age of patient

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

What species of malaria cause tertian fever?

A

Fever that recurs every third day, ie after 48hrs
Falciparum
Vivax
Ovale

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

What species of malaria cause quartan fever?

A

Fever that recurs every fourth day, ie after 72hrs
malariae

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

Basics of malaria pathophysiology

A

Erythrocytes infected with trophozoites and schizonts
Sequestration intravascularly, especially in capillaries
Rosetting (infected RBCs bind to uninfected RBCs)
Microvascular obstruction (due to RBCs becoming less deformable)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

What does pfEMP1 stand for and what does it do?

A

P falciparum erythrocyte membrane protein 1
Mediates cytoadherence
Exported to the surface of infected RBCs
Interacts with molecules on endothelial cells including ICAM-1
Different subsets determine pathophysiology and clinical features

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

What codes for pfEMP1? How does change in genes affect the protein and its capability?

A

About 60 genes called var.
Each parasite can switch on and off specific var genes -> produces functionally different protein to evade host’s immune system

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

What are the principles of immunity to malaria?

A
  • Its partial and sterile
  • gradual acquisition can result in people living in endemic areas due to repeated exposure to parasites -> they lose this when they leave
  • Two types of immunity in parallel: anti-parasite + anti-disease
  • Age and exposure independently impact on development of immunity -> kids in moderate/high transmission settings develop immunity faster as transmission increases
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

What does anti-parasite and anti-disease immunity mean?

A

Anti-parasite: ability to control parasite density
Anti-disease: ability to tolerate higher parasite densities without fever

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

Intervention that has had the biggest impact on malaria control/case prevention?

A

Insecticide treated nets

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

Why has progress stalled in malaria control?

A

— COVID-related disruptions
— Climate change
— Humanitarian crises
— Limited funding
— Insecticide & artemisinin resistance
— Invasion of An stephensi
— Health systems challenges

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

Ideas for new tools to combat malaria?

A

Vaccines
Longer lasting insecticides (non-pyrethroid? -> growing resistance)
Vector tools to address outdoor biting
Single-dose preventive therapies (monoclonal Abs)
Diagnostic tools for latent P.vivax infection
New drugs to mitigate ACT resistance
Single-dose chemoprevention therapies

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

What is the R0 or basic case reproductive rate?

A

The defined no. of secondary cases arising from a single primary case in a fully susceptible human population

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

Vectorial capacity

A

Total no. of potentially infectious bites that arises from all the mosquitoes biting a single perfectly infectious human on a single day

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

What does R0=1 mean?

A

Stable transmission
Each new case gives rise to a single further case – Total number of cases remains the same

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

R0>1 means?

A

Cases increase until entire population is infected

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

R0<1 means?

A

Cases decline until infection disappears

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

What is the Ross-MacDonald equation?

A

R0 = ma2pn/-r(logeP)

  • m = mosquito density (number of female mosquitoes per person)
  • a = the frequency with which each female bites a human (vs animal)
  • p = survival rate (probability of mosquito surviving a full day after being infected)
  • n = length of sporogonic (extrinsic) cycle
  • r = recovery rate of humans
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

What are the danger signs for malaria cited in the IMCI (integrated management of childhood illness) for children 2-59 months in areas of malaria transmission?

A

Fever or hx of fever in past 24h OR palmar pallor plus 1 or more:
- Unable to eat or drink or breastfeed
- Vomiting everything
- Multiple convulsions
- Lethargy
- Unconsciousness
- Stiff neck
- Chest indrawing or stridor

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

What are the danger signs in the IMAI (integrated management of adult illness) for older children and adults in areas of malaria transmission?

A

Fever or hx of fever in past 24h plus 1 or more:
- Very weak or unable to stand
- Convulsions
- Lethargy
- Unconsciousness
- Stiff neck
- Resp distress
- Severe abdominal pain

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

Definition of severe falciparum malaria

A

One + of below with P falciparum asexual parasitaemia in absence of alternative cause:
- Impaired conciousness
- Prostration
- Multiple convulsions
- Acidosis
- Hypoglycaemia
- Severe anaemia
- Renal impairment
- Jaundice
- Pul oedema
- Significant bleeding
- Shock
- Hyperparasitaemia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

After what diagnostic tests over what period of time can you rule out a diagnosis of malaria?

A

At least 3 diagnostic tests over 24-48hr

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

What molecules do RDTS detect for malaria?

A

Histidine rich protein 2
Plasmodium lactate dehydrogenase (pLDH)

Both can increase through trophozoite development

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

Treatment for non-severe malaria?

A

ACT: eg artemether + lumefantrine (AL), or artesunate + amodiaquine (AS + AQ)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

Reasons for recurrent malaria or patient coming back sick again?

A

Can result from re infection or recrudescence (eg treatment failure)
- ?drug resistance
- Suboptimal dosing
- Poor adherence or vomiting
- Substandard or fake medicines

Need to confirm recurrence with microscopy (not RDTs)

May not be possible to distinguish recrudescence from infection in individual patients -> need PCR for genotyping
Re infection very common in high risk areas

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

Management of treatment failure for malaria (non-severe)

A

Within 28 days: alternative ACT

After 28 days: consider as new infection and treat with first line ACT

Note: do not retreat with AS+MQ (artesunate + mefloquine) -> increased risk of neuropsychiatric reactions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
93
Q

Alternative treatment for uncomplicated falciparum malaria if ACTs are not available

A
  1. Malarone (atovaquone-proguanil), weight based, OD, 3 days
  2. Quinine + doxycycline, Q 8hrly + doxy daily, 7 days
  3. Quinine + clindamycin, Q 8hrly + clinda 8hrly, x7days
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

How to treat falciparum in pregnancy?

A

1st trimester: artemether-lumefantrine

2/3rd trimesters: treat with ACT

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

What is a key SE/risk of quinine treatment for malaria?

A

Hypoglycaemia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

What malaria drugs should never be used in pregnancy?

A

Primaquine
Tetracyclines eg doxy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

What treatment should be given to prevent relapse of vivax or ovale?

A

Primaquine (0.5mg/kg/day) for 7 days for children and adults

NOT if pregnant or breastfeeding

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

What treatment should be given to women pregnant or breast feeding if vivax or ovale? (ie to prevent relapse)

A

Weekly chemoprophylaxis with CQ until delivery and breastfeeding are completed
If then G6PD ok -> primaquine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

What treatment should be given to G6PD negative people to prevent relapse with vivax or ovale?

A

Primaquine base at 0.75mg/kg bw once a week for 8 weeks (with close medical supervision)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

41 yo woman presents to hospital with fever, chills, vomiting x 3d. She’s 18 weeks pregnant & lives in Uganda. She’s had malaria several times before but has no
other PMH. No concerning signs for severe malaria. PCR positive for P ovale. Treatment?

A

CQ - treatment dose for two days then weekly suppression whilst pregnant and breast feeding

Once stopped BF then check G6PD level and if ok -> for 7 days primaquine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

How did the evidence on primaquine change in 2022?

A

Alternative standard dose -> shortened course regimen to 0.5mg/kg/day for 7 days rather than treating for 14 days

Was made as shorter regimen could lead to better adherence and to fewer relapses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

How does primaquine work for falciparum?

A

Kills mature (stage V) gametocytes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

What treatment is recommended to reduce risk of onward transmission of falciparum malaria in endemic low transmission areas?

A

Single dose of primaquine + ACT

(Not in pregnant/BF women, infants <6mths, or older infants with unknown G6PD status)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

You are asked to provide pre-travel advice for a family traveling to East Africa on holiday to visit relatives for 3 weeks.

The following family members are planning to travel:
* Mother – aged 35, 15 weeks pregnant
* Son – aged 3 years, weight 17kg
* Daughter – aged 3 months, weight 5.2kg
* Grandmother – aged 65, history of depression

What will you recommend?

A

Mum: either mefloquine if no hx of MH issues and if grandmother is paternal, otherwise don’t go

Son: Malarone (or mefloquine)

Daughter: mefloquine

Grandmother: Malarone (or doxy)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

Non-chemoprophylaxis advice for malaria

A

– Sleep under an insecticide treated bednet,
– Cover up between dusk and dawn
– Repellants with 30-50% DEET

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

Common severe malaria complications in adults

A

Coma/convulsions
ARDS
**AKI
Shock/hypoglycaemia
Hyperparasitaemia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

Common severe malaria complications in kids

A

**Coma/convulsions
AKI
Shock/hypoglycaemia
**Severe anaemia
Respiratory distress secondary to acidosis
Hyperparasitaemia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

WHO/UK threshold for hyperparasitaemia

A

WHO: >10%
UK: >2%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

Blantyre coma score

A

Motor:
Localises painful stimulus 2
Withdraws limb from pain 1
Non-specific or absent response 0

Verbal:
Appropriate cry 2
Moan or inappropriate cry 1
None 0

Eyes:
Directed/following 1
Not directed 0

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
110
Q

Holoendemic

A

Disease for which a high prevalent level of infection begins early in life and affects most of the child population, leading to a state of equilibrium such that the adult population shows evidence of the disease much less commonly than do children

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

Hyperendemic

A

Persistent, high levels of disease occurrence

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

Mesoendemic

A

An endemic disease with a moderate rate of infection. This term is often used to describe the prevalence of malaria in a local area, with 10 to 50% of children showing evidence of prior infection being considered a moderate level for that disease.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
113
Q

Hypoendemic

A

A disease that is constantly present at a low incidence or prevalence and affects a small proportion of individuals in the area. A term commonly used in malaria literature to refer to regions with low transmission.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
114
Q

In early years eg up to 3 years old, what is the dominant severe falciparum malaria manifestation in low vs high endemic areas?

A

Areas of low transmission: cerebral malaria

Areas of high transmission: severe anaemia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

What three pathophysiological processes result in most of the major manifestations of severe falciparum?

A

Sequestration
Microvascular obstruction
Organ dysfunction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

Clinical features associated with case fatality from malaria?

A

– Impaired consciousness (multiple factors)
– Presence of retinopathy (highly suggestive)
– Convulsions common (but can be sub-clinical)
– Increased ICP (in > 80% of children, NL in adults)
– Cerebral oedema (more common in children)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

Features of retinopathy in malaria

A
  • Retinal whitening (macular or peripheral)
  • Discolouration of retinal vessels (white or orange)

Non specific:
- Retinal haemorrhages, especially with white centres
- Papilloedema

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

Levels of severe anaemia in malaria for children and adults

A

Haemoglobin < 5g/L in children, < 7g/dL in adults

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

Causes of severe anaemia in children and adults with malaria?

A

– Spleen filters both infected & damaged uninfected cells
– Intravascular haemolysis
– Bone marrow suppression
Repeated malaria infection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
120
Q

Apart from the infection itself, what are some other contributors to severe anaemia with malaria?

A

HIV, bacterial infections, hookworm, deficiency of B12 & Vitamin A

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

What drives breathing issues in malaria with kids vs adults?

A

Kids: acidosis + pneumonia

Adults: ARDS, pul oedema, pneumonia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

What is the most common pathogen associated with bacteraemia with malaria?

A

non-typhi Salmonella

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

Clinical features associated with case fatality from malaria in AQUAMAT trial?

A

Base deficit >8mmol/L
Coma
Uraemia (Bun?20mg/dL)
Underlying chronic illness

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

What are warning signs of severe malaria on peripheral smear?

A

High parasite density
Presence of schizonts on peripheral blood smear
Pigment in the neutrophils

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

What did both AQUAMAT and SEQUAMAT show?

A

Both showed in African and South Asian setting artesunate>quinine for severe malaria

SEQUAMAT showed even better in those with high parasite counts as brings counts down in 2 days whereas quinine takes 7

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

Basic management of severe malaria

A
  1. Admit
  2. IV/IM artesunate for a minimum of 24hrs (2.4mg/kg at 0,12,24 hrs then 24hrly, if <20kg then 3mg/kg)
  3. Stepdown to ACT 3 days
  4. Consider primaquine if vivax/ovale or low transmission area for falciparum

ALSO CONSIDER GIVING EMPIRICAL ABx eg CEFFTRIAXONE

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

Key side effect of artesunate?

A

Risk of delayed haemolysis

Rapidly kills ring-stage parasites which are then taken out of red cells by the spleen

These RBCs then return to circulation but with shortened life span -> haemolysis

Occur often 1 week after treatment: patient with s/s anaemia and dark urine (bilirubin)

Worse in patients with hyperparasitaemia and non-immune travellers

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

Benefit of artemether over quinine

A

Need smaller volumes for IM injection
In kids >5 and adults may be superior to quinine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

How do you manage patients pre-referral for suspected severe malaria?

A

Where complete treatment of severe malaria is not possible, but injections are available, adults and children should be given:
○ A single IM dose of artesunate & referred to an appropriate facility for further care
○ If IM artesunate is not available, give IM artemether or, if that is not available, IM quinine

Where IM injection of artesunate is not available:
○ Children < 6 years should be treated with a single rectal dose (10mg/kg bw) of artesunate & referred immediately to an appropriate facility for further care
○ Rectal artesunate should not be used in older children and adults

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

What are some of the neurological sequelae of CM and how long do they persist for?

A

Resolve within 6 mth: Hypotonia, ataxia, tremors, cortical blindness, aphasia

Persist: Motor deficits

Worsen: SAL(T) + cognitive deficits

Develop: behavioural disorders, epilepsy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
131
Q

What are some of the associations of placental malaria and which pregnancy is most at risk?

A

First pregnancy then reduction in risk -> can acquire Abs against CSA-binding iRBCs and resistance to placental malaria

IUGR
Preterm labour
LBW
SGA
Stillbirth

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

Pathophysiology of placental malaria

A
  • Sequestration in intervillous space of placenta
  • Cytoadherence via chondroitin sulphate A

First time mothers: lack immunity to CSA-binding parasites so highly susceptible to parasitaemia and chronic infection
SO important reservoir for infection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
133
Q

Why can malaria be difficult to diagnose during pregnancy?

A

Women living in endemic areas develop anti-malarial immunity from lifelong exposure to falciparum parasites -> controls infection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

What is IPTP?

A

Intermittent preventitive treatment of malaria during pregnancy.

Given:
Curative dose of an effective antimalarial drug (currently sulfadoxine-pyrimethamine) to all HIV-negative pregnant women in areas of moderate to high malaria transmission without testing whether infected or not

Usually given at each antenatal visit

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

When should a blood transfusion be considered for severe anaemia in malaria?

A

– In high transmission settings, transfuse children if Hb < 5 g/dL (Hct < 15%)
– In low transmission settings, Hb < 7 g/dL (Hct < 20%)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

What does HIV infection do to malaria severity?

A

increases malaria severity
- Higher parasite densities (in partially immune)
- Risk of severe malaria & death (in less immune)
- In pregnant women, ↑adverse effects of placental malaria

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

What are two relevant drug reactions in HIV if being managed for malaria?

A

Efavirenz increases AQ exposure & risk of toxicity

Avoid artesunate and sulfadoxine if taking co-trimoxazole

Avoid artesunate and amodiaquine if being treated with efavirenz or zidovudine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

Common severe manifestations of vivax

A

Severe anaemia
Respiratory distress (especially in children)
Acute renal failure

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

How is Knowlesi defined?

A

Defined as for falciparum but with two differences
– Hyperparasitaemia = parasite density > 100,000/µL
– Jaundice & parasite density > 20,000/µL

Associated with high parasite densities, treat as for falciparum

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
140
Q

WHO guidance for severe resistant malaria

A

Treat with artesunate and CQ together
Repeat films after course of treatment

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

Current threats to malaria control?

A

Drug resistance
Insecticide resistance (pyrethroids)
P falciparum lacking HRP2/3 (RDTs don’t pick up parasite)
Anopheles stephensi

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

Goals for TB vaccine

A

Prevent infection, progression of disease especially pulmonary, prevent reinfection and reactivation

Better TB vaccine for neonates/infants compared to BCG (aim to be safer in immunocompromised) + more sustainable to produce

Vaccine to improve TB treatment outcomes -> prevent re-infection, increase proportion of survivors to cure, shorten and simplify drug treatment regimens (ie vaccine in addition to treatment)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

Risk factors for childhood TB

A
  • Infectiousness of index case
  • Age of child
  • Proximity to and duration of contact with index cases
  • Immune status of child
  • Children of families living with HIV
  • Other illnesses eg HIV, pertussis, malnutrition, post-measles etc
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

Three most frequent symptoms of TB in children

A
  1. Persistent unremitting cough >2-3 weeks
  2. Intermittent fever
  3. Weight loss/failure to gain weight
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
145
Q

What is the differentiation between young and older kids in terms of TB presentation and diagnosis? (binary)

A

Young children <5
- High risk of LN and disseminated disease
- Clinical diagnosis more common

Kids >15 years
- Higher risk of adult type disease
- Bacteriological confirmation of TB is common

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
146
Q

Why do young children eg <2 years old have a higher risk of LN TB and disseminated disease?

A

Immaturity of immune system
Less resident macrophages and existing ones more immature

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
147
Q

Two pathways for TB infection in kids?

A
  1. 50-90% -> containment with granuloma leading to latent infection (more likely the older child is)
  2. Primary progressive pulmonary disease -> disseminated disease (military TB, TB meningitis)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
148
Q

Investigations for TB

A
  1. Immune based tests – Tuberculin Skin T est; Interferon-γ Release Assay
  2. Radiology – X ray, CT scan
  3. Sputum samples collection
  4. Pathogen detection tests:
    i. Microbiological tests – culture
    ii. Molecular / Nucleic acid amplification test – GeneXpert, Truenat
  5. Histological tests – FNAB/FNAC
  6. Biomarkers – futuristic, subject of intense research
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
149
Q

How does the TST work, what does a positive result suggest and what is it impacted by?

A

Delayed-type hypersensitivity reaction to PPD; heterogenous mix of mycobacterial proteins

§ Positive TST suggests M.tb infection or sensitization to M.tb Ag - results do NOT indicate or rule out TB disease

Impacted by prior BCG vaccination (false positive), NTM exposure, HIV

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
150
Q

Is IGRA helpful to diagnose TB in kids?

A

§ Sub-optimally sensitive in children + in immunosuppression
Indeterminate results common in young children

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
151
Q

Common CXR manifestations of TB in kids?

A

Hilar lymphadenopathy
Pulmonary effusion
Paratracheal lymphadenopathy
Miliary TB
Adult type disease if older eg dense alveolar consolidation, cavities

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
152
Q

Types of sputum sample collection for children with suspected TB?

A

Spontaneous sample: older children or adolescents
§ Sputum induction (<7 years)
§ Gastric washings (<6mths)

BAL
§ Not necessary if induction can be done
Personnel need to be trained, hospital admission needed with overnight fasting

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
153
Q

What is the value of culture for TB diagnosis in kids?

A

Childhood TB is paucobacillary: 95% of TB cases in children <12 years are smear negative

Culture-positive: 4.2% (<5yo), 5% (5-9 yo) and 21% (10-14yo)

Culture positivity in kids rarely exceeds 30%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

How are the majority of TB cases in children diagnosed?

A

Clinical features +/- radiology

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

Extrapulmonary manifestations in children of TB

A

Enlarged, non-tender LNs (esp cervical)

Painful swelling of joint or bone, which has slowly arisen over time

Angular deformity of spine

Abdominal swelling - hard, painless (+/- free fluid)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
156
Q

What are the challenges in diagnosing TB in kids?

A

1) TB in children mimics other common childhood diseases
2) Childhood TB is paucibacillary in nature
3) CXR changes often non-specific; inter-reader variability
4) TST / IGRA of limited value in TB-endemic settings

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
157
Q

CSF features of TBM

A

Elevated opening pressure
Cells (?mononuclear, can be some neutrophils, variable no.)
Elevated protein
Low glucose

Xpert ultra and culture ~50% (no single test can rule out TBM)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
158
Q

Relationship between diabetes and TB?

A

Diabetes is associated with active TB -> 3.6 fold higher TB risk

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
159
Q

What is latent vs subclinical TB?

A

Latent
- Asymptomatic
- Sputum does not contain enough bacteria -> can’t culture it
- CXR would be negative

Subclinical
- No symptoms and don’t feel ill
- But TB prevalent and active
- Ie if given a sputum pot they can produce some and TB can be grown
- Might have some CXR changes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
160
Q

What is MDR TB?

A

Resistance to rifampicin and isoniazid

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
161
Q

Pre-diagnostic loss to follow up (LTFU)

A

Screened for disease but no investigations

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
162
Q

Pre-treatment LTFU

A

Diagnosed with disease but not started on treatment

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
163
Q

Normal regimen for TB prevention?

A

Daily isoniazid
6-9 months

(most programmes go for 6 months)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
164
Q

Other options compared to standard regimen for TB prevention? 2024 update,

A

Weekly rifapentine and isoniazid for 3 months
- issues with regulation and rolling out
OR
Daily isoniazid plus rifampicin for 3 months
OR (conditional recommendation)
1 month regimen of daily rifapentine + isoniazid or 4 months of daily rifampicin

2024 update:
levofloxacin daily for 6 months for people exposed to multidrug- and rifampicin-resistant TB

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
165
Q

Normal regimen for TB prevention in children<15 years?

A

Daily rifampicin and isoniazid for 3 months

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
166
Q

What was the DOTs policy package? What disease was it for? What are the 5 strategies?

A

TB
○ Increase political will
○ Diagnosis based on quality-assured sputum testing: “passive case finding”
○ Uninterrupted standard 6 months treatment course ie uninterrupted drug supplies
○ Patient support (DOT - directly observed therapy)
○ Systematic reporting % cure

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
167
Q

What are the possible treatment outcomes for TB?

A
  • Treatment success: Cured + Treatment completed cases
  • Failed treatment
  • LTFU
  • Died
  • Not evaluated
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
168
Q

Diagnostics for Malaria

A
  1. Field based
    ○ Microscopy
    ○ RDTs
  2. Molecular methods
    ○ qPCR (quantitative PCR/real time PCR)
    ○ PCR (eg nested PCR methods)
    Genome sequencing (eg field-based Nanopore sequencing)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
169
Q

How can falciparum parasites produce a false negative RDT result?

A

Parasites found to lack HRP2 and HRP3
- Method of resistance to evade diagnostics ie gene deletion

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
170
Q

Why is vivax more widely spread geographically?

A

Can replicate/survive at lower temperatures
- Can be transmitted by lots more mosquito species
- More transmissible to mosquitoes and make them earlier on in infection
- ?day biting so nets less effective

Traditionally less efficient at invading Duffy negative RBCs but this is now changing

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
171
Q

Which malaria parasite has the fastest erythrocytic cycle?

A

Knowlesi

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
172
Q

What plasmodium can cause chronic, undetectable infection?

A

malariae
low parasitaemia
quartan fever pattern
slowest erythrocytic cycle

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
173
Q

Where is Knowlesi restricted to and why?

A

SE Asia esp Malaysia
Mosquito presence
Macaques prevalence

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
173
Q

Two species of plasmodium ovale?

A

Curtisi
Wallikeri

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
174
Q

Information to obtain from diagnostics for malaria?

A
  • Species?
  • Parasite density?
  • Is it original infection or relapse?
  • Any drug resistant genotypes?
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
175
Q

How can microscopy correlate with severity of clinical status of malaria?

A

Lots of gametocytes -> sicker patient
Parasite starts accelerating sexual reproduction for onward transmission

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
176
Q

3 formulations of RDTs for malaria?

A

○ Pan-plasmodium LDH
○ P.falciparum-specific HRP2
○ P.falciparum-specific LDH

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
177
Q

What plasmodium species gives a lot of false negatives with RDTs?

A

Ovale

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
178
Q

Can you use nested PCR to quantify parasitaemia?

A

No
Only species detection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
179
Q

What type of molecular diagnostic test can you use to work out species and parasitaemia?

A

qPCR or rtPCR

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
180
Q

What can whole genome sequencing tell you for malaria?

A

○ Species
○ Drug resistance markers
○ Multiplicity of infection (clonal/polyclonal -> ie are there multiple strains of species infecting patient)
○ Original infection/relapse
○ Origin of infection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
181
Q

Goals of the End TB strategy vs SDG targets

A
  • Drop TB cases (reduce incidence 90%)
  • Drop deaths from TB (reduce by 95%)
  • Drop catastrophic costs due to TB

By 2035

(Goals made in 2014)

SDG targets by 2030
- reduce incidence by 80%
- reduce deaths by 90%
- reduce catastrophic costs to 0

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
182
Q

TB is most prevalent in which group

A

Adult men = 55%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
183
Q

Evidence based TB interventions that could be scaled up straight away (3)

A
  1. Digital CXR + Computer Aided Diagnostics then sputum PCR at primary care
    ○ Convenience screening +/- symptoms
    CAD since 2022 being recommended for first time as alternative to human interpretation
  2. Urine-LAM (TB antigen) screening for all hospitalized HIV+ medical patients
    ○ New LAM tests may expand this remit
  3. Community interventions with evidence for affordable impact
    - Digital CXR-based
    - Symptom + smear microscopy-based
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
184
Q

Microscopic features of Plasmodium falciparum

A
  • Red cells NOT enlarged
  • Rings appear fine and delicate and there may be several in one cell
  • Some rings can have two chromatin dots
  • Presence of marginal or applique forms
  • Unusual to see developing forms in peripheral blood films
  • Gametocytes: crescent shape appearance
    ○ Do not appear in peripheral blood in first few weeks of infection
    Maurer’s dots may be present
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
185
Q

Microscopic features of Plasmodium vivax

A
  • RBCs containing parasites usually enlarged
  • Schuffer’s dots in red cells
  • Mature ring form large and coarse
  • Developing forms frequently present
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
186
Q

Features of plasmodium malariae malaria

A
  • People can be infected for long periods before symptomatic
  • Incubation 18-40 days
  • Quartan fever pattern
  • Slowest growth => longest erythrocytic cycle
  • Associated with rare kidney pathologies and fatalities
  • Frequently low parasitaemias
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
187
Q

Microscopic appearance of plasmodium malariae

A
  • Ring forms can have a squarish appearance
  • Can also be bulls eye -> ie centralised in cell
  • Trophozoites can be band form or compact
  • Mature schizonts -> daisy head appearance with 8-10 merozoites
  • Red cells NOT enlarged (parasite likes old cells)
    Chromatin dot may be on inner surface of ring
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
188
Q

Microscopic features of Plasmodium ovale

A
  • Red cells enlarged
  • Comet forms common
  • Rings large and coarse
  • Schuffner’s dots
  • Can have fimbriae -> jagged edge
  • Mature schizonts
  • Similar to those of P malariae but larger and more coarse
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
189
Q

Which plasmodium species shows seasonal variability?

A

Ovale

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
190
Q

Causes of eosinophilia

A

Atopic
Haematological
Rheumatological
Drugs
Infection - most commonly parasites

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
191
Q

Differentials for transient, migratory lesions

A

Loa loa
Gnathostoma
Sparganum

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
192
Q

Why should you not use ivermectin to treat loa loa and what should you use instead?

A

Can result in large inflammatory reaction in people who have lots of microfilariae eg encephalopathy due to body reaction in response to worms being killed

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
193
Q

What is a dimorphic fungi?

A

Yeast in the beast
Mould in the cold
eg histoplasma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
194
Q

Treatment of causes of transient migratory lesions

A

Albendazole or Ivermectin (but not IVM for loa loa)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
195
Q

Clinical tests for fungi

A

Beta D glucan
Galactomannan
Serology (less helpful in immunocompromised patients eg HIV, as can’t mount a proper response)
If particular suspected cause, then specific test eg Histoplasma urinary Ag

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
196
Q

Treatment of disseminated histoplasmosis

A

Amphotericin B (ambisome is liposomal form)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
197
Q

Treatment of localised, pulmonary histoplasmosis

A

Itraconazole

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
198
Q

Examples of dimorphic fungi

A

Histoplasma
Blastomyces
Coccidioides
Paracoccidiodes
Sporothrix
Penicillum = Talaromyces

Containment level 3

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
199
Q

Two common yeast species causing disease in humans

A

Candida eg albicans or non-albicans
Cryptococcus neoformans

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
200
Q

What type of organism does India ink reveal thick capsule for?

A

Cryptococcus neoformans

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
201
Q

What disease might a person pick up by inhaling air in a bat cave?

A

Histoplasmosis
Common in bat faeces

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
202
Q

Manifestations of disseminated histoplasmosis

A

Reticuloendothelial involvement
Skin nodules
Mouth ulcers
Renal, bone, CNS, liver lesions
Adrenal involvement (hypo)
Uveitis, panophthalmitis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
203
Q

A farmer from rural Sudan presents with 6 months of worsening foot pain, non-healing ulcers and difficulty walking. USS showed a soft tissue mass behind the ankle and he had some nodular skin changes over the heal. What is most likely diagnosis and how would you treat?

A

Madura foot eg Eumycetoma pedis
Prolonged course of antimicrobial eg Itraconazole or Voriconazole

  • Difficult as need levels and monitoring
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
204
Q

Triad of mycetoma eg Madura foot

A

Tumour - growth
Tracts - body’s attempt to drain and get pus out
Grains - release of grainy liquid of fungal colonies

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
205
Q

Most common cause of fungal Madura foot

A

Madurella mycetomasis

  • a environmental soil mould
  • hot spot in Sudan
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
206
Q

Differentials for mycetoma ie Madura foot

A

Actinomycosis
Podoconiosis
TB
Non-tuberculosis mycobacterium
Bacterial osteomyelitis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
207
Q

Types of liver flukes

A

Fasciola (big)
Opisthrochis
Clonorchis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
208
Q

Treatment for fasciola

A

Triclabendazole 10mg/kg/PO
2 doses 12-24 hours apart

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
209
Q

What are opisthrochis and clonorchis and where are they endemic? How do you get them? How do you treat them? What pathology are they associated with long term?

A

Liver flukes eg trematodes
Central and east Asia eg Kazakhstan, Siberia, eastern Russia

Eating raw or undercooked fish

Treat with praziquantel

Cholangiocarcinoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
210
Q

How do you get fasciola hepatica or gigantica?

A

Eating watercress

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
211
Q

How can you diagnose fasciola hepatica acutely vs chronically?

A

Acute infection
§ Eosinophilia
§ May have positive serology (6-8 weeks for Ab to develop)
§ Negative OCP (not early enough to get eggs in stool)

Chronic
§ Positive serology
§ May have eosinophilia
§ Intermittent eggs in stool

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
212
Q

Differentials for liver abscess?

A

Bacterial
Amoebic (singular, big, ring-enhancing)
Echinococcosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
213
Q

Differentials for umbilicated lesions in SE Asia

A
  • Molluscum contagiosum
  • Cytomegalovirus
  • Cryptococcus neoforms
  • Dimorphic fungi: histoplasmosis/talaromycosis
  • Syphilis
  • TB
  • Trichoepitheliomas
  • Tuberous sclerosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
214
Q

28M from Northern Thailand presents with Fever + weight loss +
non-productive cough + skin lesions + abdominal discomfort.
O/E: hepatomegaly and he is covered in umbilicated lesions. A biopsy shows yeast with inner septum. What is the diagnosis and treatment?

A

Talaromycosis caused by Talaromyces marneffei

Management:
- Severe:
○ Induction: Ambisome 7-14 days or Amphotericin B IV (0.7mg/kg/day) at least 14 days
○ Consolidation therapy: Itraconazole PO BD for next 10 weeks

Mild/moderate:
Itraconazole

If HIV positive then secondary prophylaxis with itraconazole until CD4+ count of >100 cells for at least 6 months (suggestion)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
215
Q

Features of talaromycosis

A

Dimorphic fungus
Bamboo rats are reservoir
Associated with HIV and poverty
Infectious mould spores inhaled from soil
Skin lesions only present in around 50% of patients (usually late stage infection)

Symptoms: weight loss, prolonged fever, fatigue, diarrhoea
Signs: hepatosplenomegaly, anaemia, lymphadenopathy

CXR: chest signs common, may see diffuse reticulonodular or alveolar infiltrates

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
216
Q

Relationship between incidence, duration of HIV and prevalence

A

P ~/= ID

Treatment increases -> duration of disease to rise + incidence to fall
- HIV prevalence should continue to rise or be stable for time to come
Means need to consider its role in aetiology and management of other conditions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
217
Q

When were ARTs first approved by US FDA?

A

1987, more in 1990s and 2000s

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
218
Q

The HIV fast track targets (by 2030)

A

95 95 95
* First 95: 95% of PLHIV know their status
* Second 95: 95% of PLHIV on treatment
* Third 95: 95% of PLHIV on treatment achieved viral suppression

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
219
Q

Where is HIV incidence now increasing?

A

Middle income countries eg
- West Africa
- South America
- Asia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
220
Q

Where is HIV-2 most prevalent?

A

West Africa and places with ties to WA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
221
Q

Mechanisms of transmission of HIV

A

Sexual
- Transmission through semen, vaginal fluid, blood, anal mucus
- Heterosexual transmission is main mode of spread globally

Parenteral
- Transmission through skin or via blood/blood products

Mother-child transmission
During pregnancy, labour or through breastfeeding

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
222
Q

HIV prevention tools

A
  • Treatment
    ○ Treatment as prevention (U=U)
  • HIV combination prevention
    ○ Behaviour change including consistent condom use
    ○ Biomedical interventions: eg Voluntary medical male circumcision, Pre-exposure prophylaxis (PrEP)
    ○ Supportive political and health care environment
  • Prevention of mother-to-child transmission (PMTCT)
    HIV testing for access to treatment and some prevention
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
223
Q

Equation for HIV reproductive number

A

R0 = c x ß x D

R0: Basic reproductive number
C: Exposure rate
–Influencing factors: Norms around multiple partnerships, sexual violence

ß: Probability of HIV transmission
–Influenced by: Ability to negotiate condom use, age at first sex, availability of PrEP

D: Duration of infectious period
–Influenced by: Access to treatment

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
224
Q

What is a concentrated HIV epidemic?

A

HIV has spread rapidly in one or more defined subpopulation but is not well established in the general population.
Numerical proxy: HIV prevalence is consistently over 5% in at least one defined subpopulation but is less than 1% among pregnant women in urban areas.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
225
Q

What is a generalised HIV epidemic?

A

HIV is firmly established in the general population.
- Numerical proxy: HIV prevalence consistently exceeding 1% among pregnant women. Most generalized HIV epidemics are mixed in nature, in which certain (key) subpopulations are disproportionately affected.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
226
Q

What is a mixed HIV epidemic?

A

People are acquiring HIV infection in one or more subpopulations and in the general population. Mixed epidemics are therefore one or more concentrated epidemics within a generalized epidemic.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
227
Q

WHO identified populations at risk of HIV infection?

A

Five key populations in 2016 WHO guidelines
1. Sex workers*
2. People who inject drugs
3. Men who have sex with men (MSM)
4. People in prisons or other closed settings
5. Transgender people

Also region- or country-specific populations at elevated risk
- Migrant populations
- Occupational groups: fisherfolk, truck drivers, miners
Adolescents*

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
228
Q

Why are adolescent girls at higher risk of HIV?

A
  • Biological vulnerability: higher per-act risk of sexual transmission, higher STI prevalence, anatomical development
  • Behavioural vulnerability, including risky sexual behaviour
  • Social and structural vulnerability: gender-based violence, lack of access to services increase risk and decrease access to PrEP and other HIV prevention
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
229
Q

Properties of HIV (virology)

A
  • Retroviridae family
  • Lentivirus
  • HIV-1 and HIV-2 species
    Single stranded, sense RNA
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
230
Q

Process of HIV infection (cellular)

A
  • Gp41 and Gp120 bind onto CD4 cell using CCR5 or CXCR4 co-receptor
  • Fusion with membrane
  • Expels capsid into cell cytoplasm
  • Capsid makes way to nucleus via nuclear import (using microtubules)
  • Then viral RNA exposed
  • Reverse transcription
  • Integration of proviral DNA into human genome -> stays there forever in that cell
  • Transcription to make proteins eg enzymes, viral particles
  • Nuclear export
  • Assembly of new virus with utilisation of cell membrane -> budding
  • Release of new virion
    Maturation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
231
Q

Pathogenesis of HIV

A
  • Local infection of mucosal tissues
    ○ Can’t detect virus in blood in this stage
  • Death of mucosal memory CD4+ T cells
  • Infection established in lymphoid tissues eg LNs
  • Viraemia after infection in blood
  • Spread through body
  • Immune response -> but is incomplete and dysfunction ie partially control viral replication
    ○ Abs can’t completely neutralise virus
    ○ Cytotoxic T lymphocytes
  • Due to widespread dissemination of HIV in lymphoid tissue -> clinical latency due to reservoir
    ○ Chronic + concentrated low-level viral production in lymphoid tissues
    ○ Inflammation associated with viraemia
    § Cytokine storms
    § CV disease
    § Metabolic issues
  • Eventually increased viral replication
  • AIDs -> destruction of lymphoid tissues + depletion of CD4+ T cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
232
Q

How to test for HIV? What are the respective window periods?

A

Two tests: 4/5th generation assays
○ Ie get two positive results
Then immunotyping

4/5th generation include:
p24Ag + Abs combined OR
separate p24 Ag and Ab components
(p24 is protein in viral caspid)

Window period 45 days for Ab/Ag assays, 90 for POCT

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
233
Q

What does high viraemia in HIV mean for HIV species?

A
  • Immune selection pressure –> escape mutants –> quasispecies created
  • 1-10 mutations per replication cycle, with production of 1 billion virions a day
  • Older infection = more diverse
  • Drug selection pressure
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
234
Q

What is virological failure in context of HIV?

A
  • Failure of ART to reduce viral load
    WHO can define as viral load >1000 copies on ideally two tests
  • Need to do genotypic resistance test:
    Protease and integrase are standard

Can result from archive mutations -> means can re-emerge if patient re-started on treatment having been incompletely treated in the past (which may be unknown to healthcare provider)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
235
Q

What is pre-treatment drug resistance in context of HIV?

A

HIVDR detected in individuals initiating ART regardless of prior antiretroviral (ARV) drug exposure(s) (e.g. prior ART followed by default from care ie stopped taking it or lost to f/u, pre‐exposure prophylaxis (PrEP), post‐exposure prophylaxis (PEP))

Means archive mutations can re-emerge when ART is started

IE: PDR may either be transmitted at time of initial infection or acquired by prior exposure to ARV drugs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
236
Q

Drug resistance concepts for HIV

A
  • Mixed population including drug-resistant minority variants
  • Subtherapeutic drug levels e.g. adherence issues
  • Selection pressure -> resistant variants flourish
  • Usually not “fitter” than wild-type
  • Take away drug (ART cessation/onward transmission)
    ○ wild-type takes over again
  • Archived resistance
  • Compartmentalisation e.g. CNS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
237
Q

Properties of HIV-2

A
  • Origin West Africa
  • More indolent
  • Slower progression
  • Slower CD4 decline
  • VL lower
  • Intrinsic NNRTI resistance
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
238
Q

What is the most important determinant of OI risk?

A

CD4 count at baseline

Two components:
1. Number of CD4 T cells
2. Differences in phenotype and function of pathogen-specific CD4 T cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
239
Q

What is the definition of advanced HIV disease?

A

CD4<200 cells/mm3
OR
WHO stage 3 or 4 event

Children<5 years old with HIV

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
240
Q

Clinical manifestations of HIV?

A
  1. Primary HIV infection
    ○ Acute viral illness or asymptomatic
  2. Chronic viral infection: direct viral effects and inflammation
    ○ Adenopathy, haematological, skin disorders
    ○ Higher risk of non-HIV cancers, cardiovascular disease, osteoporosis
    ○ HAND, wasting syndrome, HIVAN
  3. Opportunistic infections and cancers: impaired cellular immunity
    ○ Many pathogens; related to CD4 count
  4. ART-associated effects eg IRIS, toxicity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
241
Q

What is IRIS?

A

Immune reconstitution syndrome
- High mortality in first 12 months of starting ART
- Usually 4-6 weeks after starting ART

Two forms:
○ Paradoxical IRIS
○ Unmasking IRIS

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
242
Q

Causes of lymphadenopathy in HIV at any CD4 count

A

TB (TB - IRIS)
Syphilis
Lymphoma
KS/Castlemans
NTM

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
243
Q

Causes of lymphadenopathy in HIV at CD4<100

A

Nocardia
Dimorphic fungi infection eg hits, emergomycosis, talaromycosis, sporotrichosis
Cryptococcus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
244
Q

Mucocutaneous manifestations of mild/early HIV infection

A
  1. Herpes zoster
  2. Papular pruritic eruption (PPE)
  3. Molluscum contagiosum
  4. Apthous ulcer
  5. Oropharyngeal candidiasis
  6. Oral hairy leukoplakia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
245
Q

What is PPE in HIV and how does it present?

A

Papular pruritic eruption
○ Eosinophilic folliculitis
○ Occurs at any CD4 count
○ ++ discomfort; can lead to scarring
○ No specific treatment:
§ Antihistamines, symptomatic
○ Easily confused with dimorphic fungal infection –> always biopsy if CD4 <100 or constitutional symptoms

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
246
Q

Differentials for molluscum like rash in HIV?

A

§ Cryptococcosis
§ Histoplasmosis, talaromycosis
§ Mpox

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
247
Q

Causes of apthous ulcers

A

§ Early HIV
§ Lymphoma and KS
§ Herpesviruses: HSV, CMV
§ Other infections
§ Tuberculosis
§ Syphilis
§ Deep fungal infection
§ Leishmaniasis
§ Rheumatologic diseases and IBD

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
248
Q

Two forms of pneumocystis jiroveci and characteristics? How do you diagnose PCP?

A

Ascus forms (cystic)
Transmissible form
Thick cell wall containing beta-glucans
Contains cholesterol (not ergosterol, so can’t be targeted by traditional anti-fungals)
Immune stimulus

Trophic forms (trophozoite)
Replicative form (more abundant in lungs)
Wall-less -> biofilm
Contains cholesterol (not ergosterol, so can’t be targeted by traditional anti-fungals)
Immune evasive

DIAGNOSING IT”
§ Gold standard bronchoscopy: not available
§ Sputum-based tests not accurate enough
§ Most patients started on empiric therapy

qPCR:
good rule out test on induced sputum and oral wash

B-D-Glucan:
After a negative test: Rules out PCP with 95% certainty up to a pre-test probability of 50%

After a positive test:
Increases chance of PCP to ~70% for any HIV-
associated pneumonia (prevalence of PCP ~25%)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
249
Q

Treatment for PCP

A

Co-trimoxazole 21 days high dose

TMP-SMX = trimethoprim-sulfamethoxazole, used both for PCP prophylaxis and treatment

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
250
Q

Respiratory illnesses at any CD4 count in HIV

A

Bacteria (especially pneumococcal disease)
TB and PTLD
Resp viruses
NTM

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
251
Q

Respiratory illnesses in HIV with CD4 <200

A

PCP
KS
Lymphoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
252
Q

Respiratory illnesses in HIV with CD4 <100

A

Cryptococcus
Dimorphic fungi
CMV

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
253
Q

What causes oral cell leukoplakia? How can you differentiate it from candidiasis?

A

EBV
- No treatment

You can do tongue scrapings on candida throat/tongue, not on OCL

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
254
Q

Differential diagnosis for mediastinal lymphadenopathy in HIV

A

TB
Lymphoma
Mycobacteria
Dimorphic fungi
Cryptococcus
Nocardia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
255
Q

Differential diagnosis for pleural effusion in HIV

A
  • TB
  • Bacterial
  • KS
  • Cryptococcosis
  • Lymphoma (KSHV-related)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
256
Q

Treatment for toxoplasmosis?

A

Treat toxoplasmosis with co-trimoxazole

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
257
Q

Differential diagnosis of lymphocytic meningitis in HIV

A
  • TBM
  • Cryptococcal meningitis
  • Viral meningitis
  • Bacterial meningitis (pneumococcus and meningococcus)
  • Listeria
  • Syphilis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
258
Q

Differential diagnosis of intracranial mass lesion in HIV

A
  • TB
  • Toxoplasmosis
  • Nocardia
  • Pyogenic brain abscess
  • Lymphoma
  • Cryptococcus
  • Syphilis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
259
Q

Differentials for severe weight loss in HIV

A
  1. Opportunistic diseases - TB, NTM, lymphoma, deep fungal, leishmaniasis
  2. HIV wasting syndrome
  3. GI pathology
    - Painful oral/oesophageal lesions
    - Severe chronic diarrhoea
  4. Other malignancy
  5. Depression – very common & often unrecognized
  6. Lack of financial resources to buy food
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
260
Q

35 years old female
Known HIV positive, treatment defaulter, CD4 = 14 cells/µL
1 seizure
2 week history of headache
Stiff neck
Cough for 1 month
Weight loss, diarrhoea
Mild meningism
Glasgow Coma Score 14/15

Ddx?

A

Toxoplasmosis
CMV
PML
Cryptococcal meningitis
TBM
Bacterial meningitis
Viral meningitis
VZV
Primary CNS lymphoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
261
Q

Pathogen detection tests for CNS infections in HIV

A

○ Stains (Gram, AFB, India Ink)
○ Bacterial, mycobacterial and fungal culture
○ Cryptococcal antigen (CrAg) test (close to 100% sensitivity and specificity)
○ Xpert Ultra MTB/Rif
○ Pneumococcal Ag tests
○ Viral and bacterial PCRs (multiplex)
○ Toxoplasma serology

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
262
Q

Non-specific investigations for CNS infections in HIV

A
  • Blood tests
    ○ Malaria film, FBC, U&E
    ○ Blood cultures
    ○ HIV test and CD4 count
  • LP
    ○ Opening pressure
    ○ Appearance
    ○ Cell count & differential
    ○ Protein & glucose

Only real contraindication to LP in LMICs where scanning is not possible is focal neurological deficit (excluding CN palsies) suggesting mass lesions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
263
Q

How is cryptococcal meningitis investigated?

A

CrAg tests (close to 100% sensitivity and specificity), india ink staining (ring enhancing), culture

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
264
Q

What did the Ambition-cm trial show?

A

Single high-dose liposomal amphotericin based regimen for treatment of HIV-associated Cryptococcal meningitis

○ Novel single dose treatment was both safer and more effective
Now being used in Botswana, Eswatini, Malawi, Zimbabwe, Uganda

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
265
Q

Treatment of cryptococcal meningitis

A

Induction treatment:
Single-dose Liposomal Amphotericin B* 10mg/kg plus
Flucytosine (5FC)100mg/kg/day (4 divided doses) & Fluconazole 1200mg/day for 2 weeks; then

Consolidation treatment:
Fluconazole 800mg/day for 8 weeks; then

Maintenance treatment:
Fluconazole 200mg/day until CD4 > 200 cells/µL and virally suppressed on ART

  • Do not give steroids -> worsen outcomes
    Start ART 4-6 weeks after starting anti fungal to avoid IRIS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
266
Q

Clinical manifestations of Amphotericin B toxicity and management?

A
  • Renal impairment, K+/Mg2+ wasting, anaemia, thrombophlebitis
  • Give 1L N. Saline + 20mEq K+, 2 Slow-K Tablets (16 mEq) twice daily, 2 Slow-Mag Tablets (143mg Mg2+)
    Alternate day creatinine + electrolytes, twice weekly Hb
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
267
Q

Should you give steroids in cryptococcal meningitis?

A

No - worsens outcomes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
268
Q

When should you start ART for cryptococcal meningitis?

A

Start ART at 4-6 weeks after initiating antifungal therapy to avoid immune reconstitution syndrome

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
269
Q

How common is raised ICP in cryptococcal meningitis? How do you manage it?

A

Common ≈ 75 %, can occur at initial presentation, on treatment, IRIS

Communicating hydrocephalus

Headache, Visual/hearing loss, CN palsies, depressed GCS, confusion

Management is with therapeutic lumbar punctures - daily LP’s until OP <25cm H20

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
270
Q

Who should be screened for cryptococcus? If positive how should they be managed?

A

Individuals with CD4<100

Ideally offer LP if available

Treat patients without evidence of CNS disease with high dose fluconazole 1200mg for 2 weeks, then standard consolidation and maintenance therapy

If LP positive then manage as cryptococcal meningitis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
271
Q

When should you consider TBM in a patient?

A
  • Chronic headache + meningism
  • Cryptococcal investigation negative
  • CSF compatible
  • Routine bacterial cultures negative
  • Evidence of TB elsewhere (TB on CXR: 44% in HIV+ TBM)

Often CD4 count may be >100-200

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
272
Q

Management of TBM

A
  • Standard quadruple therapy (RIPE), 9-12 months duration
  • Steroids: maybe but benefit in HIV-infected subset not clear - currently evidence suggesting no benefit nor in TB pericardial disease
    -ART: data on timing not clear, most would delay initiation until around 9-12 months after initiation of TB treatment and give steroid cover

TB medication may help prevent TBM in HIV positive individuals. Give BCG in kids.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
273
Q

CD4 level likely in toxoplasmosis in HIV+ people?

A

CD4 <100cells/microL

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
274
Q

Management of toxoplasmosis

A

Initial therapy:
- Pyrimethamine 200mg OD loading then 75mg OD plus Sulfadiazine 6-8g/daily divided QDS
Or
- Clindamycin 450mg pd QDS

Increasingly used:
Co-trimoxazole (high dose) much more widely available

Secondary prophylaxis:
- Initiate ART without delay (low risk of IRIS)
Long term co-trimoxazole prophylaxis until sustained (>6m) response to ART with CD4 count >200 cells/µl

Don’t give steroids -> can confuse therapeutic trial

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
275
Q

Top HIV associated malignancies

A

Cervical cancer
Kaposi’s sarcoma
Lymphomas esp Non-hodgkin’s lymphoma
Castlemann’s disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
276
Q

Clinical features of TB in individuals without HIV vs HIV infected immunosuppressed individuals

A
  • Pulmonary TB
  • Upper lobe infiltrates with cavities
  • EPTB less common
  • Mild immunosuppression (eg CD4 >350) is similar

VS

  • Thoracic lymphadenopathy and pleural effusions more common
  • Extrapulmonary disease more common with declining CD4 counts
  • Disseminated disease
    • Post-mortem have shown dissemination in 90%
  • Lower zone nodules, non-cavitating or normal CXRs
  • Very severe:
    • Sepsis with bacteraemia + organ dysfunction
    • ~25% admitted with sepsis have TB. Microbacteraemia in 40-50%
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
277
Q

Key microbiological investigations for TB

A

Microscopy - sputum smear for AFB
Culture - gold standard
Molecular techniques
- PCR eg GeneXpert
Antigen detection eg urinary LAM (only can use in HIV)
Can do urine Xpert Ultra (high specificity but lower sensitivity) and stool Xpert Ultra

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
278
Q

4 TB screening options for patients with HIV

A
  1. WHO 4 symptom screen (cough, fever, weight loss, night sweats)
  2. CRP, cut-off >5 mg/L
  3. Chest X-ray (+CAD)
  4. WHO recommended molecular diagnostic (medical inpatients)

Screen with IGRA or TSTs
Screen at every visit to a health facility

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
279
Q

What features in adults living with HIV should prompt TB testing and what tests?

A

PLHIV
-Signs /symptoms of TB
-Advanced HIV
-Seriously ill
-Positive screening test

Do:
- urinary LAM
- Sputum Low complexity molecular testing eg geneXpert

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
280
Q

What features in children living with HIV should prompt TB testing and what tests?

A

-Signs /symptoms of TB
-Positive screening test

Do
Respiratory sample -> Low complexity molecular testing eg geneXpert
Stool
Low complexity molecular testing eg stool Xpert ultra

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
281
Q

What is Focused Assessment with Sonography (FASH) for HIV/TB ?

A

Use USS as POC to look at 6 points to look for:
- Pleural fluid
- Pericardial fluid
- Ascites
- Focal liver lesions
- Focal splenic lesions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
282
Q

When should ART be started in patients with HIV and TB? What is the exception?

A

Start ART as soon as possible within two weeks of initiating TB treatment regardless of CD4 cell count among people living with HIV

Exception: TBM (start 4-8 weeks after)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
283
Q

What is paradoxical TB-IRIS?

A
  • Started TB treatment and newly go on to start ART
  • Then clinical deterioration
  • Usually happens within weeks of ART
    Paradoxical as patients should get better and not worse
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
284
Q

What is unmasking TB-IRIS?

A
  • Not on TB treatment
  • Newly started on ART
  • After a couple of weeks -> deteriorate and new S/S + new diagnosis of HIV associated TB
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
285
Q

Management of TB in HIV?

A

RI for 6 months
PE for 2 months

+ pyridoxine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
286
Q

Common SEs of TB treatment?

A

Hepatitis
Rash
Arthropathy (pyrazinamide)
Optic neuritis (ethambutol)
Peripheral neuropathy (isoniazid)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
287
Q

What is the issue with co-treating TB and HIV? What regimens are suggested for it?

A

Rifampicin - potent CYP450
Many HIV drugs metabolised by CYP450

  1. Atripla: efavirenz + lamivudine + tenofovir
    ○ But issues with resistance
  2. Dolutegravir + Tenofovir + lamivudine + dolutegravir 50mg
    ○ Ie give with rifampicin but need to double dose of dolutegravir so give extra tablet top up
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
288
Q

Management of TB IRIS?

A

Steroids eg PO Prednisolone

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
289
Q

Features of TB IRIS

A
  • 8% have paradoxical IRIS after starting ART
  • Usually worsening pulmonary infiltrates / enlarging LNs
  • 25% hospitalized, 2% mortality
  • Prednisolone reduces severity /hospitalization / need for procedures

Risk factors:
○ Low CD4 count / high viral load
○ Extrapulmonary TB
○ Early ART (but mortality lower)
○ Prophylactic prednisolone reduces IRIS by 30%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
290
Q

Risk factors for TB IRIS

A

○ Low CD4 count / high viral load
○ Extrapulmonary TB
○ Early ART (but mortality lower)
○ Prophylactic prednisolone reduces IRIS by 30%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
291
Q

Parasite factors involved in malaria clinical outcome

A

Drug resistance
Multiplication rate
Invasion pathways
Cytoadherence
Rosetting
Antigenic polymorphism
Antigenic variation via PfEMP-1
Malaria toxin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
292
Q

Host factors involved in malaria clinical outcome

A

Immunity
Proinflammatory cytokines
Genetics eg SCT, thalassaemia, ovalocytosis, CD36, TNF-a, ICAM-1 etc
Age
Pregnancy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
293
Q

Geographical and social factors involved in malaria outcome

A

Access to treatment
Cultural and economic factors
Political stability
Transmission intensity -> seasonality, infectious bites per year, epidemics

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
294
Q

What processes actually cause fever in malaria?

A

Schizont rupture
- release of parasite proteins eg immunogenicity materials triggering immune response

Malaria toxins
eg Haemozoin, Parasite DNA, GPI (Glycophosphatidylinositol)
causes pro-inflammatory cytokine release

Splenic clearance of iRBC via activation of splenic macrophages -> cytokine storm -> systemic inflammation over activation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
295
Q

What are the two major hypothesis for cause of CM?

A
  1. Mechanical hypothesis = vascular obstruction by parasitised erythrocytes
    ○ Evolutionary basis of cytoadherence to brain endothelial cells
    § To avoid clearance of parasitised erythrocytes by the spleen
    § For optimal development condition: low O2 pressure found in post-capillary venules
  2. Immunopathology hypothesis = effector function of immune cells
    ○ Endothelium activation and damaging
    § Up-regulation of parasite, platelets and monocytes receptors (cell accumulation within microvessels)
    § Endothelial shrinking, induction of apoptosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
296
Q

What host endothelial receptor is important for cytoadherence in falciparum malaria?

A

CD36
- Parasites sequester in microvessels via CD36 -> avoid clearance by the spleen

○ CD36 is a scavenger host endothelial cell receptor which is widely distributed

Over 75% of encoded PfEMP1s have a binding domain for this receptor

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
297
Q

3 processes through which sequestration of falciparum malaria occurs

A
  • Cytoadherence
  • Rosetting
  • Clumping

Impair blood flow leading to anaerobic glycolysis, acidosis and tissue damage

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
298
Q

What cytokine levels correlate with mortality, parasitaemia and disease severity in CM?

A

TNF-alpha

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
299
Q

What are the two main complications of pregnancy associated malaria?

A
  1. Preterm delivery (<37wks)
    ○ Associated with malaria parasitaemia and anaemia
  2. Foetal growth restriction
    ○ Chronic infection and placental insufficiency
    ○ Lack of nutrients to foetus

○ TNF, IFN-Y, IL-1 and IL-2 production elevated
§ High TNF linked to fetal growth restriction and preterm delivery
IFN-Y related to low birth weight

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
300
Q

What is the pathophysiological of placental malaria?

A
  • iRBCs accumulate and aggregregate in placental intervillous spaces
  • RBC use the VAR2CSA type of PfEMP-1, which binds to chondroitin sulfate A (CSA)
  • The sequestration blocks the transfer of nutrients from mother to embryo
  • VAR2CSA found during pregnancy that are absent in men and non-pregnant women
  • Women with PAM remain afebrile - suggesting local pathology - with some aspects of acquired immunity remaining unaffected by placental parasitisation
  • Elevated type 1 pro-inflammatory responses in placentas of malaria-infected mothers
  • Sequestration of infected erythrocytes -> inflammatory environment -> release of C&C -> recruitment of monocytes and macrophages into intervillous space

○ TNF, IFN-Y, IL-1 and IL-2 production elevated
§ High TNF linked to fetal growth restriction and preterm delivery
IFN-Y related to low birth weight

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
301
Q

Two unique features of placental malaria

A
  • VAR2CSA found during pregnancy that are absent in men and non-pregnant women
  • Women with PAM remain afebrile - suggesting local pathology - with some aspects of acquired immunity remaining unaffected by placental parasitisation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
302
Q

Pathophysiology of malarial anaemia

A
  1. Loss of infected RBCs: lysis or phagocytosis
    ○ Haemolytic anaemia

○ Misconception: SMA arises simply from the lysis of infected RBC. ➔ Lysis of infected RBCs is not the most significant cause of SMA

○ Minor loss, parasitaemia < 1%

  1. Loss of uninfected RBCs
    ○ Phagocytosis, phosphatidylserine and other markers involved as ‘eat me’ signal
    ○ ~12 uninfected RBC are lost for every iRBC during malaria infection
  2. Suppression of BM erythropoiesis -> prevention of replacement
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
303
Q

Why are uninfected RBCs destroyed during malarial anaemia?

A

Changes in plasma membrane of uninfected RBCs during malaria infection targets them for destruction: ’eat me’ signal
* Phosphatidylserine externalization
* Oxidation
* Reduced deformability

Binding of immune components to the surface of uninfected RBCs also targets cells for destruction
* Binding of malaria RSP-2 (ring surface protein 2) on the surface facilitates recognition
* Immune complexes and IgG (specific for parasite material that may stick to uninfected RBC) promotes phagocytosis
* Complement binding

Destruction of uninfected RBCs generally mediated by splenic red pulp macrophages

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
304
Q

How is erythropoiesis suppressed during severe malarial anaemia?

A
  • Haemozoin (a breakdown product of haemoglobin produced by the parasite) inhibits erythropoiesis
  • Suppressive cytokines:
    ○ TNF inhibits all stages of erythropoiesis
    ○ IFN-Y inhibits erythroid cell growth, differentiation and survival
  • Hepcidin is released by the liver and blocks iron availability for incorporation into haemoglobin
  • MIF (migration inhibitory factor) inhibits erythropoiesis by direct effects on erythroid precursors and bone marrow rather than modulation of TNF production
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
305
Q

What do erythrocytes containing altered Hb display less of in malaria?

How do haemoglobinopathies confer an advantage to prevent/dampen malaria pathogenesis?

A

PfEMP1

  • Prevention of parasite from establishing its own actin cytoskeleton within the host cell
  • Impaired vesicular transport
  • Distorted trafficking of PfEMP1; less in the surface
  • Reduced cytoadherence
  • Data limited to HbS and HbC
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
306
Q

What is reservoir of leptospirosis?

A

Comes from proximal tubules of mammals

so high in animal urine eg rats
outbreaks when floods

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
307
Q

What are the four types of epidemic curves and their principles?

A

Point source:
- Questions typically refer to weddings eg outbreak of vomiting after cream puffs at a wedding
- Should settle after exposure is over
- Most cases within same incubation period

Extended common-source:
- The continual exposure means continue cases
- If you stop the exposure, ie break the water pump, then the cases should stop

Propagated source:
- Transmission occurs from person to person rather than a common source
- Can have multiple waves
- Cases in one peak can be sources for cases in a subsequent peak
- May have progressively taller peaks (if incubation period and infectiousness period similar)

Intermittent source
- Multiple peaks
- Length bears no relation to incubation period -> reflects intermittent times of exposure
- eg contaminated food source

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
308
Q

What are the 12 steps for outbreak responses?

A
  1. Prepare for field work -> identify an investigation team, mobilise appropriate resources
  2. Establish the existence of an outbreak/epidemic
  3. Verify diagnoses of cases
  4. Establish a working case definition
  5. Systematic case finding to identify additional cases
  6. Conduct descriptive epidemiological studies (time, place, person)
  7. Develop hypotheses
  8. Evaluate hypotheses (often with case control study)
  9. If required, reconsider/refine hypotheses and do additional studies to test them
  10. Implement control and prevention measures (as early as possible)
  11. Communicate findings
  12. Institute surveillance
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
309
Q

Three types of case definition in outbreaks?

A
  • Proven/confirmed
    ○ Usually are symptomatic
    ○ Are within suspected incubation period
    ○ Have positive test: ie serology, culture, PCR etc
  • Probable/suspected
    ○ ?live in epidemic area
    ○ Symptomatic
  • Possible
    ○ Milder symptoms
    ○ ?Not necessarily in traditional incubation period
    ○ ?Might have had contact with proven or probable case in preceding given amount of time
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
310
Q

Two forms of protozoa, which one causes infection?

A

Cysts (can sit/lay dormant for a while)
Trophozoites (cause active infection)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
311
Q

Non infectious causes of bloody diarrhoea

A
  • IBD
  • Ischaemic colitis
  • Radiation colitis
  • Bowel cancer
  • Vasculitis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
312
Q

Viral causes of bloody diarrhoea

A

VHF esp Lassa, Marburg, ebola
-CMV if CD4 low

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
313
Q

Top bacterial causes of bloody diarrhoea

A
  • Shigella (p to p spread)
  • E coli (p to p spread)
  • Campylobacter
  • Salmonella

Other
- C difficle
- Yersinia enterocolitia
- Plesiomonas shigelloides

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
314
Q

Parasitic causes of bloody diarrhoea

A
  • Schistosomiasis
    ○ Intestinal -> eggs in mesenteric veins -> portal spread -> portal HTN and varices
    § Can distinguish from other liver diseases as liver function quite preserved
  • Amoeba (E histolytica)
  • Balantidium coli
  • Malaria
  • Helminths
  • Very heavy trichuris (but tends to be due to rectal prolapse)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
315
Q

Electrolyte deficiency with invasive shigellosis?

A

Hypokalaemia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
316
Q

Precautions for faecal-oral spread

A

PPE
Hand-washing
Sanitation
Incineration
Clean surfaces
Identify common sources for spread
Prevent iatrogenic infections

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
317
Q

What are the drug classes for HIV treatment?

A

NRTI: nucleoside reverse transcriptase inhibitors
- Pretend to be nucleotides
- Drug class associated with most toxicities - ?secondary to introduction of them into cell
- Risk of mitochondrial toxicity

NNRTI: non-nucleoside reverse transcriptase inhibitors
- Bind to reverse transcriptase and cause confirmational change so doesn’t work as well

INSTI: integrase (strand transfer) inhibitors

PI: protease inhibitors

Capsid inhibitor (lenacapavir): 6 monthly subcut injection, not yet licensed, could be gamechanger

Other drugs that aren’t really used:
CD4 inhibitor: ibalizumab
- Withdrawn in Europe, only available in USA
- IV every 2 weeks
- Very expensive

CCR5 inhibitor: maraviroc
- Rarely used

Fusion inhibitor: enfuvirtide
BD subcut injections

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
318
Q

When should rapid ART for HIV NOT be started?

A
  • TBM: after at least 4 weeks, but start within 8 weeks of anti-TB treatment
    ○ Consider adjuvant steroids
  • After TB treatment started: within 2 weeks
  • Cryptococcal meningitis
    ○ Increased mortality with immediate ART
    ○ Defer until 4-6 weeks after CM treatment start
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
319
Q

Until 2000s what ART formula would HIV patients have?

A

Until 1996: NRTI

1996 - 1999: PI + NNRTI

2000+: INSTI + new (2 NRTI + 1 other)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
320
Q

What is the characteristic resistance mutation to Lamivudine?

A

M184V

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
321
Q

What is the only effective dual drug therapy for HIV ART?

A

Dolutegravir (II)
Lamivudine (NRTI)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
322
Q

WHO guidelines for ART?

A

First line:
Tenofovir-DF (TDF) + emtricitabine or lamivudine + dolutegravir
(TLD = Teno/Lamni/Dolu is 1 pill but not available in UK yet)

Alternative first line:
TDF + lamivudine + efavirenz 400

Don’t use efavirenz if country has resistance rates of >10%
if DTG not available then boosted PI should be used
Consider TAF instead of TDF if pre-existing bone disease or renal impairment

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
323
Q

What are TDF or TDX and what are their biggest SEs?

What could you start instead?

A

Tenofovir disoproxil fumarate = old tenofovir = TDF, TDX (same drug but different salts)
- These are old tenofovir

Biggest SEs: renal impairment and reduction in bone mineral density

Can consider new tenofovir: TAF = Tenofovir-alafenamide

or another drug

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
324
Q

If an individual is established on ART, what test should be used to monitor treatment according to WHO guidelines?

A

Monitor viral load
Don’t monitor CD4 count

Now in London/?UK once CD4>350 stop monitoring it

If viral load testing not available then use CD4 cell count with clinical monitoring

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
325
Q

What threshold can be used to assess ART failure using dry blood spot specimens?

A

> 1000 copies/ml
Same as for plasma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
326
Q

What biochemical marker can dolutegravir cause a harmless rise in?

A

Creatinine
Not indicative of renal disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
327
Q

37 year old man
Chronic HBV 04/2022 starts
tenofovir-DF + lamivudine + efavirenz FDC

10/2023: 6M viral load undetectable
04.2024: 12M viral load 2700

What would you do?

A

Resistance testing if available
Otherwise, according to WHO guidelines if Viral load >1000 on NNRTI then switch as pre-treatment resistance common in many countries

Switch to 2nd line after single VL >1000 reduces mortality &
transmission & is cost-effective (at $500 DALY threshold)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
328
Q

Key toxic drug reaction examples with ritonavir or cobicistat? (4)

A

§ Midazolam
§ Amiodarone
§ Rivaroxaban
§ Fluticasone

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
329
Q

British HIV guidelines for treatment?

A

INSTI + 2NRTIS
- TAF + emtricitabine + bictegravir (Biktarvy)
- Abacavir/lamivudine/dolutegravir (if HLAB5701 negative, 10-year CV risk <10%)
- Dolutegravir + TAF/TDF + lamivudine or emtricitabine

OR

INSTI + 1 NRTI
- Dolutegravir + lamivudine
○ No baseline lamivudine resistance
○ Baseline VL <500,000
○ Baseline CD4>200
○ No need to include tenofovir for hep B treatment/prevention

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
330
Q

What intervention can reduce the risk of transmission of HIV by 50%?

A

Male circumcision

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
331
Q

FTC for HIV - full name?

A

Emtricitabine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
332
Q

TDF for HIV - full name?

A

Tenofovir disoproxil fumarate

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
333
Q

TAF for HIV - full name?

A

Tenofovir-alafenamide

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
334
Q

RAL for HIV - full name?

A

Raltegravir

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
335
Q

What does post-exposure prophylaxis for HIV involve?

A
  • Up to 72 hour after exposure
  • POCT negative
  • Tenofovir/emtricitabine and raltegravir OD for 28 days
  • Vaccinate for hepatitis/assess risk for other STIs
  • Emergency pregnancy prevention
  • Follow up testing for HIV
    ○ Minimum 45 days after completion of PEP course
    ○ If 28 day PEP course is completed this is 73 days (10.5 weeks) post-exposure
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
336
Q

What does pre-exposure prophylaxis for HIV involve?

A
  • Truvada OD (can be on it long term) = FTC/TDF (for anyone at risk) OR Descovy (FTC/TAF) OD for men and transgender women
  • Cabotegravir IM injection every 8 weeks
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
337
Q

Population most at risk of HIV at the moment?

A

Cisgender adolescent and young women

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
338
Q

Brand name for emtricitabine and tenofovir disoproxil?

A

Truvada

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
339
Q

What ARTs is Descovy?

A

Emtricitabine/tenofovir alafenamide

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
340
Q

What monitoring should be done for patients on ART?

A

VL: at 6M, 12M then yearly
CD4 every 6M until established on ART

Ideally also check
Creat & eGFR for TDF
Pregnancy test for women of child bearing potential

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
341
Q

What monitoring should be done for patients with suspected ART failure?

A

Creat & eGFR for TDF

HBV surface Ag before switching drugs if not done at baseline or negative and not-vaccinated

If >50-<1000 copies/ml VL: enhanced adherence counselling and repeat VL testing at 3 months -> if above >1000 then switch, if <50 then maintain, IF still >50-<1000 copies/ml VL then maintain ART, enhanced adherence counselling and repeat VL testing at 3 months -> if still high then switch

If VL>1000copies/ml and on NNRTI based ART -> switch
(high risk of resistance)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
342
Q

A patient is started on ART and on yearly check VL is 750 copies/ml, what do you do?

A

Enhanced adherence counselling and repeat VL testing at 3 months

IF still >50-<1000 copies/ml VL then maintain ART, enhanced adherence counselling and repeat VL testing at 3 months -> if still high then switch, if <50 then maintain

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
343
Q

How does dolutegravir cause a rise in creatinine?

A

Inhibits OCT2, a renal transporter
Highest in first 4 weeks
Not harmful to kidneys nor reflective of renal disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
344
Q

What is Hy’s law?

A

Jaundice + transaminitis on ART -> associated with worse mortality

Switch the most likely culprit

○ Eg AZT to TDF
○ 3rd agent to another 3rd agent
○ Atazanavir = PI
§ Not really used anymore
§ Can cause jaundice
§ Risk of renal impairment

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
345
Q

A patient with HIV on atazanavir has hyperbilirubinaemia on routine blood tests, would you switch their ART?

A

Only if jaundiced + transaminitis
Atazanavir alone causes unconjugated hyperbilirubinemia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
346
Q

What is the WHO screening guidance for patients with HIV for Hep B and C?

A
  • At enrolment to care/initiation of ART
  • At treatment failure/switching ART regimen
  • At re-engaging following care interruption
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
347
Q

WHO recommendations for NNRTI resistance?

A

Switch away from NNRTI at 1st detectable VL
- Pre-treatment resistance common in many countries
- Resistance common at virological failure
- Although many will resuppress, outweighed by risk of more resistance + consequences of treatment failure
- Switch to 2nd line after single VL >1000 reduces mortality & transmission & is cost-effective (at $500 DALY threshold)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
348
Q

WHO guidance for second line ART regimens?

A

If non-DTG regimen is failing:
- DTG in combination with optimised NRTI backbone

If DTG-based regimens are failing:
- Boosted PIs with optimised NRTI backbone

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
349
Q

2 types of causative agents for hookworm and examples:

A
  1. Human/intestinal hookworm
    ○ Humans are definitive hosts
    § Ancyclostoma duodenale
    § Necator americanus
  2. Animal hookworm
    ○ Humans are accidental host
    ○ Multiple species
    ○ Can invade and parasitise humans
    ○ Ancyclostoma ceylanicum
    ○ Ancyclostoma braziliense
    ○ Ancyclostoma caninum
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
350
Q

Features of cutaneous larva migrans

A

Hookworm infection
- ‘Skin-worm-moving’
- Intensely pruritic
- Creeps along for days “serpinginous”
- Track – due to an inflammatory, allergic response to the microscopic larva
- Represents IgE-mediated response to filariform larval migration → histamine release → itchy ++
- Secondary bacteria infection can occur
- Gradually will self-resolve once larvae enter circulation and migrate to the lungs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
351
Q

Features of lung migration of hookworm?

A
  • Usually asymptomatic
  • Less commonly → eosinophilic pneumonitis
  • IgE-mediated immune response to larvae in the lungs
  • Leads to transient cough, wheezing, chest pain and haemoptysis
  • Lobar infiltrate on a CXR can mimic bronchopneumonia
  • “Loeffler’s syndrome”
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
352
Q

10 key facts about strongyloidiasis

A

1) Widespread distribution
2) Walking barefoot on soil contaminated with human faeces
3) Mostly asymptomatic or low grade GI disturbance
4) Auto-infection cycle for decades
5) Serpiginous rash of larva currens
6) Beware small bowel obstruction (duodenal aspirate)
7) Hyper-infection syndrome, normal eosinophils
8) Cellular immuno-suppression including HTLV-1
9) Travellers and migrants are different
10) Eosinophilia, serology, microscopy and charcoal culture

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
353
Q

What soil transmitted helminth can cause auto infection?

A

Strongyloides

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
354
Q

Classes of dimorphic fungi

A

○ Histoplasma
○ Talaromyces = penicillium
○ Blastomyces
○ Coccidioides
○ Paracoccidioides
○ Sporothrix (rose gardening one)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
355
Q

What is the 3rd most common opportunistic infection with HIV in China, Vietnam and Thailand?

A

Talaromycosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
356
Q

Management of talaromycosis

A
  • Severe:
    ○ Induction: Ambisome (ie liposome amphotericin) 3-5mg/kg/day IV for 10-14 days OR Amphotericin B IV (0.7mg/kg/day) at least 14 days
    ○ Consolidation therapy: Itraconazole PO BD for next 10 weeks
  • Mild/moderate:
    ○ Itraconazole 200mg BD for 8-10 weeks
  • Maintenance therapy:
    Itraconazole 200mg OD lifelong or until CD4>100 for 6 months while on ART
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
357
Q

What pathogen causes Talaromycosis? What are some key facts and treatment for it?

A

Talaromyces marneffei
Thermally dimorphic geographically restricted fungi
* Rainy seasons soil exposure (inhalation of conidia)
* Bamboo rats enzootic reservoir
* 3rd commonest opportunistic infection in advanced HIV in endemic areas (SE Asia- Thailand, Vietnam and China)
* Mortality 97-100% without treatment
* Delay in diagnosis doubles mortality 24-50%
* Newer diagnostics: PCR, metagenomic next-generation sequencing

Severe: liposomal Amphotericin B or amphotericin B for 14 days
Then consolidation: itraconazole BD for 10 weeks

Mild: itraconazole BD for 8-10 weeks

Maintenance: itraconazole lifelong or until CD4>100 for 6 months on ART

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
358
Q

Examples of NRTIs

A

Zidovudine (AZT), Lamivudine (3TC), Abacavir (ABC), Tenofovir disoproxil fumarate (TDF), emtricitabine (FTC)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
359
Q

Examples of NNRTIs

A
  • Efavirenz (EFV)
  • Rilpivirine (RPV)
  • Doravirine (DOR)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
360
Q

Examples of INSTI

A
  • Dolutegravir (DAG)
  • Raltegravir (RAL)
  • Bictegravir (BIC)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
361
Q

Examples of protease inhibitors

A
  • Ritonavir (RTV)
  • Lopinavir (LPV)
  • Atazanavir (ATV)
  • Darunavir (DRV)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
362
Q

What is lenacapavir? What is it used to treat?

A

Caspid inhibitor
Shown promise for HIV -> only given 6monthly subcut injections but not licensed yet

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
363
Q

In a patient with HIV who has cryptococcal meningitis when should you start ART?

A

4-6 weeks after CM treatment has started due to increased mortality with immediate ART

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
364
Q

In a patient with HIV who has TB meningitis when should you start ART?

A

After at least 4 weeks but within 8 weeks of anti-TB treatment and consider adjuvant steroids

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
365
Q

What did the START study for HIV show?

A
  • RCT on starting ART
  • Delayed (until CD4>350 or AIDS related symptoms) vs start immediate group
    Better for immediate group
  • faster viral suppression
  • lower mortality
  • lower AIDs related events
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
366
Q

What does a dolutegravir and lamivudine combination not cover for?

A

Hep B = Need tenofovir

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
367
Q

Which ART can be given IM every two months and what class does it belong to?

A

Cabotegravir

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
368
Q

A 37 year old man with chronic HBV 04/2022 starts tenofovir-DF + lamivudine + dolutegravir FDC. 10/23 VL undetectable. 4/24 VL 2,700. What do you do?

A

WHO: enhanced adherence counselling + repeat VL at 3M
* Many people will resuppress
* Resistance emergence uncommon
07/2024 VL undetectable
Consistent with data from ADVANCE (1st line DTG vs EFV) which shows most participants resuppress on continued DTG

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
369
Q

How do you encourage adherence in HIV treatment?

A

Adherence:
- Secure drug supplies
- Longer drug supplies
- Education
- Simple dosing
- Adherence tools
- Aim for longer acting medication

Attendance:
- COVID lessions
- Virtual care
- Transport support
- REACH study

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
370
Q

What did the NADIA trial show for HIV?

A
  • NADIA trial looked at dolutegravir or darunvair in combination with lamivudine plus either zidovudine or tenofovir for second line treatment of HIV
  • Showed dolutegravir is non-inferior to darunavir but is at greater risk of resistance in second line therapy
    Also tenofovir should be continued in second-line therapy rather than being switched to zidovudine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
371
Q

What are the four classes of protozoa?

A

Amoebae
Flagellates
Sporozoa
Ciliates

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
372
Q

What are the two types of amoebae to know?

A

Entamoeba
Naegleria

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
373
Q

What are the three types of flagellates to know?

A

Leishmania
Giardia
Trichomonas

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
374
Q

What are the two types of sporozoa to know?

A

Plasmodium
Toxoplasma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
375
Q

What is a type of ciliate?

A

Balantidium

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
376
Q

What are the three classes of helminths?

A

Cestodes (tapeworm)
Trematodes (flukes)
Nematodes (roundworm)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
377
Q

What are key types of cestodes/tapeworms?

A

Taenia saginata
Taenia solium
Hymenolepsis nana
Diphyllobothrium latum
Echinococcus sp

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
378
Q

What are the key types of trematodes/flukes?

A

Liver flukes eg Fasciola, Clonorchis, Opisthorchis
Lung flukes
Tissue flukes
Blood flukes eg schistosomiasis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
379
Q

What are the two types of nematodes/roundworms?

A

Geohelminths
Filariae

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
380
Q

What are types of geohelminths?

A

Ascaris
Trichuris
Hookworm
Strongyloides
Toxocara
Trichenella
Enterobius

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
381
Q

What are types of filariae?

A

Lymphatic filariasis
Loa loa
Onchocerca
Dracunculus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
382
Q

What are the key features of Ascaris?

A

“Giant roundworm”
Crenelated eggs
Adult worms 200-350mm in size
Large bowel location
Soil to mouth

Clinical features:
- Asymptomatic
Loeffler’s
GI obstruction
Cholangitis
Peritonitis

Distinct feature: lung migration

Eosinophilia only if in lungs

Treatment: albendazole

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
383
Q

What is Loeffler’s syndrome?

A

Disease in which eosinophils accumulate in the lung in response to a parasitic infection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
384
Q

What are the key features of Trichuris?

A

“Whipworm”
Bulb-ended eggs
Adult worms 50mm in size
Large bowel location
Soil to mouth

Clinical features:
Asymptomatic
GI upset
Stunting
Rectal prolapse

Distinct feature: overlap with ascaris

Often eosinophilia

Treatment: albendazole

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
385
Q

What are the key features of Ancylostoma or Necator?

A

“Hookworm”
Thin-walled eggs
Adult worms 10mm in size
Small bowel location
Skin penetration

Clinical features:
Asymptomatic
CLM
Loeffler’s
GI upset
Anaemia
VERY ITCHY

Distinct feature: lung migration

Eosinophilia can be high

Treatment: albendazole or ivermectin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
386
Q

What are the key features of Strongyloides?

A

“Threadworm in USA”
Ellipsoid eggs with larva
Adult worms 1mm in size
Small bowel location
Skin penetration

Clinical features:
Asymptomatic
Larva currens
GI upset
Hyper-infection

Distinct feature: auto-infection

Eosinophils common but not in hyper infection

Treatment: Ivermectin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
387
Q

What are the key features of Enterobius?

A

Pinworm (Or called threadworm in the UK)
Eggs are flat on one side
Adult worms 1-2mm in size
Located in rectum
Transmitted faecal to mouth

Clinical features:
Asymptomatic
Pruritis
Vaginal discharge

Distinct feature: Family clusters/sellotape test

Eosinophilia: unusual

Treatment: albendazole

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
388
Q

Basics of clinical presentation of hookworm?

A

Early illness:
- Larva migration in the skin -> cutaneous larva migrans
- Larva migration in the lungs -> coughing, wheeze

Late illness:
- Correlates with worm burden in the GI tract
- Adults bite on intestine and suck blood -> anaemia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
389
Q

Features of GI infection with hookworm

A
  • Often asymptomatic
  • Adults rarely pass spontaneously - worms hold on tight and only 1cm long
  • Two adults sit and sexually reproduce in GI tract + blood suck -> can last for years
  • Nausea, abdominal pain + diarrhoea
  • Late infections
  • Hookworm anaemia -> major concern in endemic areas
  • Each adult: 0.03-0.2ml blood/day
  • May contribute to growth restriction in heavily-infected children
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
390
Q

Lifecycle of human/intestinal hookworm?

A

Eggs in faeces
Rhabditiform larva hatches
Development to filariform larva in environment
Filariform larva penetrates skin
Larvae can be developmentally arrested and dormant in tissues
Reactivated larvae may enter the small intestine
If they get into lung circulation -> exit -> coughed up and swallowed
Eggs in faeces

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
391
Q

Life cycle of animal hookworm/cutaneous larva migrans? eg
Ancylostoma canium/braziliense
Uncinaria stenocephala

A

Skin penetration via larvae through skin
BUT animals definitive hosts
Adults in small intestine
Eggs in faeces of animal definitive host
Hatched rhabditiform larva develops in environment
Rhabditiform larva develops into infectious filariform larva

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
392
Q

Traditional story/presentation of Ancylostoma braziliense?

A

Hookworm
Person lies on beach that dogs have been running on etc (ie there faeces have been in contact with beach)
Get rash wherever their faeces touched -> clinical diagnosis of cutaneous larva migrans

Treat with ivermectin or albendazole

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
393
Q

Life cycle of ascaris?

A

Hands carry infective ova from soil contained with human excreta/dust/veg etc
Larvae penetrate mucosa
Enter lymphatics and venules
Migration to right heart and lungs -> into alveoli -> moult twice
Ascend resp tree -> descend oesophagus to mature in intestine
Maturation in humans 2 months
Adults live around 1 year and make 100,000 eggs a day

Eggs leave body and complete cycle -> have to complete in soil before affecting the next person

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
394
Q

Management of ascaris?

A
  1. Kill the worms
    ○ Albendazole
    ○ Mebendazole
    ○ Pyrantel pamoate
  2. Manage the complications
    ○ Surgery for obstruction
    ○ ERCP for pancreatitis
  3. Break the cycle
    ○ Better sanitation
    ○ Hand hygiene
    ○ MDA
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
395
Q

Clinical features of ascaris?

A

Early infection:
- Asymptomatic
Can get lung migration and Loeffler’s syndrome -> IgE mediated response to larvae in lungs -> transient wheezing, coughing, chest pain, haemoptysis
May see Charcot Leyden crystals on microscopy

Late infection:
GI worm burden related -> can be asymptomatic
Nausea/abdominal pain/diarrhoea
Malnutrition
GI complications eg bowel infection /perforation -> peritonitis

Ectopic infection -> migration up biliary tree causing pancreatitis, biliary sepsis and peritonitis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
396
Q

What parasite causes cutaneous larva currens?

A

Strongyloides

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
397
Q

What is the clinical syndrome of strongyloides?

A
  • Usually asymptomatic
  • Larval invasion of the GI tract → Abdominal pain, intermittent diarrhoea
  • Larval penetration → petechial haemorrhages and pruritis at site of entry (eg peri-anal skin)
  • Larval migration → Rash → cutaneous larva currens
  • Larval invasion of the lungs → cough, wheezing, chronic bronchitis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
398
Q

Life cycle of strongyloides

A

Filariform larvae in soil penetrate skin
Larvae move via venous system into lungs
Larvae swallowed and develop into adult females in GI tract -> eggs produced by asexual reproduction
Eggs hatch into rhabditiform larvae -> excreted in stool
Some rhabditiform larvae transform to filariform larvae and reinvade gut wall -> reinfection
The rhabditiform larvae outside of the body develop and mate ie adult males and females reproduce sexually and develop into filariform larvae

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
399
Q

What is the rash caused by strongyloides? What are some features?

A

Cutaneous larva currens
- Intensely pruritic
- Moves linearly for hours (faster moving, shorter duration than hookworm’s cutaneous larva migrans)
- Represents IgE-mediated response to filariform larval migration
- Resolves quickly when larvae enter circulation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
400
Q

Features of larval infection with strongyloides in the lung?

A
  • Usually asymptomatic
  • Less commonly → eosinophilic pneumonitis
  • IgE-mediated immune response to larvae in the lungs
  • Transient cough, wheezing, chest pain and haemoptysis
  • Lobar infiltrate on chest x-ray may mimic bronchopneumonia
    Ie “Loeffler’s”
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
401
Q

What is strongyloides hyper infection?

A

· Immunosuppressed individuals – T-lymphocyte depletion
○ HTLV-1, HIV
○ Malnourished
○ Iatrogenic immunosuppression (steroids, chemotherapy, transplant patients etc)

· Leads to a faster reproduction cycle
· Migration to ectopic sites
· Spread of faecal bacteria to normally sterile sites
· Can → recurrent gram negative bacteremia, CNS involvement, granulomas, abscesses…

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
402
Q

How should you investigate strongyloides?

A
  • Usually eosinophilia - up to 80%
    ○ Check over a time period as can have periods of high and of normal -> depends on movement of worm
  • Faecal ova, cysts and parasites – but low sensitivity – worms are difficult to detect as usually few in number. NPV 50%
  • Serology (ELISA IgG) if immunocompetent
    ○ Can stay positive for long time -> don’t use it as indicator of cure but can use it to diagnose in chronic infection
    ○ Cross reactivity for other parasitic diseases eg schiso
  • Test to do:
    ○ Charcoal stool but not routine

In hyper-infection:
- Suspect the diagnosis and try to find the worms
- Gram negative rod sepsis, bacteraemia, pneumonia, meningitis
- Eosinophilia - rare
- Serology rarely helpful -> slow turnaround, poor NPV
- Look for larvae in stool and the sputum

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
403
Q

Treatment for Strongyloides?

A

Ivermectin>albendazole
Screen and treat before planned immunosuppression (ie if risk factors then before chemo/radio etc)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
404
Q

Life cycle of trichuris?

A

Whipworm
Embryonated eggs are ingested
larvae hatch in small intestine
adults move to caecum
unembryonated eggs pass in faeces
eggs develop in soil -> become embryonated

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
405
Q

Clinical presentation of whipworm?

A

eg trichuris

Severity = correlates with burden of worms
Light infections:
○ Abdo pains

Heavy infections (800+ worms):
○ Dysentry
○ Growth stunting
○ Associated with anaemia (0.005ml blood/worm/day, unlikely causal, likely multifactorial)
○ Rectal prolapse due to tenesmus
Lots of inflammation -> higher intraabdominal pressure -> need to open bowels a lot

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
406
Q

Diagnosis and treatment of trichuris?

A

Eggs in stool OCP? Adults are visible on colonoscopy

Treat with albendazole > mebendazole

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
407
Q

What is a feature of strongyloides hyper-infection on a blood count?

A

Normal eosinophil count

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
408
Q

What is different about travellers and migrants with strongyloides infection?

A

Migrants more likely to have positive serology but negative symptoms
Travellers less likely to have symptoms but positive serology

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
409
Q

What is different about travellers and migrants with strongyloides infection?

A

Migrants more likely to have positive serology but negative symptoms
Travellers less likely to have symptoms but positive serology

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
410
Q

Lady goes to pit latrine in Nigeria late at night and gets sudden eye pain. Goes to hospital next day and eye is red with extensive corneal scarring. Cause?

A

Venom ophthalmia
Caused by spitting cobras

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
411
Q

Management of venom ophthalmia

A
  1. Immediate irrigation with any clean bland fluid
  2. Pain killing eye drops: 1% adrenaline or tetracaine (LA)
  3. Fluorescein staining/slit-lamp examination to exclude corneal abrasion
  4. Topical Abx eg tetracycline/chloramphenical
  5. Topical cycloplegic eg atropine

+/- eye pads

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
412
Q

Complications of cobra ophthalmia

A

Acute conjunctivitis
Hypopyon
Panopthalmitis
Anterior uveitis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
413
Q

Clinical manifestation of spitting cobra bites?

A

Skip lesions
Venom comes out of front of fangs
Can inject venom into lymphatics

Leads to extensive scarring, can need extensive debridement and can end up with contractures

Also can get superimposed staph infection on top or marjolin’s ulcer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
414
Q

Causes of growing burden of NCDs worldwide?

A
  • Growing and ageing populations
  • Success in preventing and treating communicable disease
  • Success in preventing childhood disease
  • Urbanisation and globalisation
  • Change in RFs eg ETOH, tobacco, diet, physical activity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
415
Q

What is the WHO: “25x25” strategy (2012)? How was this updated?

A

Aim for 25% reduction in mortality by 2025
Four priority diseases
1. CVD
2. Diabetes
3. Cancer
4. Chronic respiratory diseases

Update: 5. Mental and neurological conditions

Five priority interventions:
1. Tobacco control
2. Salt reduction
3. Improved diets and physical activity
4. Reduction in hazardous alcohol intake
5. Essential drugs and technologies

Update: address air pollution

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
416
Q

What are the 5 priorities to address NCDs in fight for UHC?

A
  1. Prevention
  2. Primary healthcare
  3. Equitable access to medicines
  4. Sustainable financing and investments
  5. Engaged and empowered communities
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
417
Q

Strategies for CVD prevention and management in LMICS?

A

Acute and chronic CVD mx
Clinical based RF control, smoking cessation, BP control
Lipid and diabetes mx
Improving medical education
Healthcare financing
Policies for smoking, diet, physical activity modulation
Tackle social determinants of health

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
418
Q

HEARTS global initiate for CVD diseases

A

Healthy lifestyle counselling
Evidence based protocols
Access to essential medicines
Risk based CVD management
Team based care
Systems for monitoring

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
419
Q

How does diabetes present differently in Black Africans compared to White Europeans?

A
  • Black Africans have lower BMI (eg 27) and lower age (51.5) + less co-existing hypertension and more hyperglycaemia
    HbA1c 9% vs 7.1% (in white Europeans)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
420
Q

WHO essential medicines for diabetes

A

Insulin
- Insulin injection
- Intermediate acting insulin
- Long-acting insulin analogues

Oral hypoglycaemic
- Metformin
- Empagliflozin
- Gliclazide

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
421
Q

Types of insulin

A

Analogue
- Rapid or long acting

Soluble
- Regular
- Short
- SC or IV

Isophane
- NPH insulin (Neutral protamine Hagedorn)
- Intermediate acting

Lente insulin
- Zinc suspension
Intermediate - long mode of action

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
422
Q

Theories behind diabetes prevalence in LMICS?

A

Thrifty phenotype diagnosis

Also lots of high sugar food/processed etc in LMICs eg South Africa

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
423
Q

Types of insulin over time they act and examples?

A

Rapid acting:
- Insulin lispro
- Aspart
- Glulisine

Short acting:
- Regular insulin

Intermediate acting:
- NPH insulin (Neutral protamine Hagedorn)

Long acting:
- Insulin glargine
- Insulin detemir

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
424
Q

Interaction between DTG and metformin?

A

Dolutegravir can increase conc of metformin by 200 times

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
425
Q

What did the INTE-AFRICA Trial show?

A

Blood Pressure: The integrated care model showed a significant increase in the proportion of patients achieving target blood pressure levels compared to standard care.

Glycemic Control: There was a notable improvement in HbA1c levels among diabetic patients in the integrated care group.

Quality of Life: Patients reported higher quality of life scores in the integrated care model.

Adherence: Higher adherence rates were observed in the integrated care group, indicating better management and follow-up.

Hospitalization: There was a reduction in hospitalization rates for chronic disease-related complications.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
426
Q

What are the obstacles to effectively managing and preventing HTN?

A

Low awareness and access to screening
Unaffordable drugs, lack of willingness/motivation to engage with services, social norms
Lack of retention due to lack of clinicians, poor understanding of conditions, fear of side effects/long term effects and competing priorities
Low screening and inaccurate measurements
Poor use of guidelines/understanding of diagnosis and management amongst healthcare staff

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
427
Q

Risk factors for stroke in LMICs

A
  • Untreated HTN
  • Infection
    ○ HIV (southern and central Africa), stroke mimics/CNS infections
  • SCD
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
428
Q

Stroke mimics in LMICs in HIV positive patients?

A

SOL
- Tuberculoma
- Cryptococcoma
- Primary CNS lymphoma
HIV associated Ois
- Toxoplasmosis
- PML
Meningitis complications
- Acute bacterial meningitis
- TB meningitis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
429
Q

Relationship between infection and stroke?

A

Infection:
- Risk of stroke, 1-3 days higher risk but can persist for 90 days
- Pre-existing risk increased in infections

Remodelling:
- Especially in HIV
- Inflammatory process or latent infection causing vasculopathies eg Moyamoya or RSCV

Vasculitis:
- Eg TB/VZV/Syphilis
- Vasotropism -> attacking of blood vessels and wall inflammation
- Endarteritis obliterans
- Post-infective inflammatory vasculitis

Hypercoagulability
- Eg COVID

Cardioembolic
- RHD
- Chagas

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
430
Q

Stroke mimics in LMICs in HIV negative patients?

A
  • Hypoglycaemia
  • Migraine
  • Todd’s paresis
  • Non-HIV associated CNS malignancies
  • Autoimmune/inflammatory CNS lesions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
431
Q

How to manage strokes in LMICs?

A
  • Investigations:
    ○ brain imaging, electrocardiogram, echocardiogram, blood test (FBC, U&Es, LFTs, CRP, HIV, VDRL), Lumbar puncture if indicated
  • Medications
    ○ low-cost medication available for management of complications
    ○ Aspirin
    ○ Insulin/anti-diabetics
    ○ Antihypertensives
    ○ Common oral agents : thiazide, amlodipine, nifedipine, atenolol, enalapril
    ○ Common IV agents: hydralazine, labetalol
    ○ Antibiotics/antivirals
    ○ Variable availability: thrombolytic agents (+/- access to confirmatory imaging)
  • Rehabilitation
    ○ Physios usually available but few staff may not meet patient needs
    ○ Other rehab specialties + equipment may not be available
    ○ Limited resource for long term / intensive rehab
    ○ Limited capacity for providing professional care and home f/u
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
432
Q

Definition of a seizure?

A

A seizure is a sudden burst of electrical activity in the brain that can affect how a person behaves, moves, or senses things for a short time.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
433
Q

Definition of epilepsy and types?

A

A chronic neurological disorder characterized by a predisposition to recurrent, unprovoked seizures.
Key features of epilepsy include:
· Recurrent seizures: A person is diagnosed with epilepsy after having two or more unprovoked seizures that occur at least 24 hours apart.
· Unprovoked: Seizures are not triggered by immediate, dentifiable causes such as fever, alcohol withdrawal, or acute brain injury.

Varied seizure types: Seizures can vary in type and severity. They may involve convulsions (tonic-clonic seizures), brief periods of altered awareness (absence seizures) or focal neurological symptoms (focal seizures).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
434
Q

Definition of status epilepticus?

A

A neurological emergency characterized by prolonged or repeated seizures without the person regaining consciousness between episodes. It is defined by either of the following criteria:
1. Continuous seizure activity lasting 5 minutes or longer without interruption.
2. Two or more discrete seizures between which there is incomplete recovery of consciousness.

Status epilepticus can be divided into two main types:
1. Convulsive Status Epilepticus (CSE): The more common and severe form, where there are continuous tonic-clonic seizures, which can lead to brain injury if untreated.
2. Non-Convulsive Status Epilepticus (NCSE): Characterized by altered mental status and seizure activity without the obvious physical convulsions.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
435
Q

Key drug management of status

A

Diazepam
Phenobarbital
Phenytoin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
436
Q

Relationship between HIV and epilepsy?

A

· Potential interactions between antiretroviral therapy for HIV and antiepileptic drugs
· Greatest concern relates to the P450 system enzyme induction effects of several older-generation antiepileptic drugs (e.g. phenobarbital, carbamazepine, phenytoin), which might lower the effective dose of non-nucleotide reverse transcriptase inhibitors (NNRTIs) and protease inhibitors (PIs), metabolized by the P450 system.
· Consequences include treatment resistance for HIV-infected patients; alternative treatment for seizures needs to be used in epilepsy patients with HIV

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
437
Q

What is the lifecycle of a mosquito?

A
  • Adult female mosquito mates soon after emergence from pupa and stores spermathecae (the sperm from male mosquitoes) to fertilise batches of eggs.
    • After the female mosquito blood feeds, egg development starts
    • Female searches for appropriate quality of water for egg laying
    • Different species of mosquito make different choices as where to lay their eggs
    • Larvae may hatch soon after eggs are laid or after some time depending on conditions
    • 4th instar metamorphoses into pupa
    • Pupa swims and breathes are but do not feed
    • Pupa metamorphoses into adult mosquito which emerges at the water surface, stretches its wings and flies away
    • Both sexes of adult obtain energy from sugary plant juices
      Only the adult female feeds on blood to get proteins to feed her young
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
438
Q

What are the two subfamilies mosquitoes are divided into?

A
  1. Anophelinae
  2. Culicinae
  3. Toxorhynchitinae (don’t like humans)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
439
Q

What diseases do not come out of Latin America?

A

Never
- Japanese
- Loa Loa
- Lassa, Ebola
- HAT
- Schistosoma haematobium

Very rarely
- Onchocerciasis
- Schistosoma mansoni

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
440
Q

Why can you get Schistosomiasis mansoni but not Schistosomiasis haematobium or japonicum in Latin America?

A

Parts of Latin America have Biomphalaria snail for mansoni but no snail for haematobium (bulinus)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
441
Q

Some diseases that are quite specific to Latin America?

A
  • Trypanosoma cruzi
    ○ esp in HIV -> causes neurological disease that looks like toxoplasmosis
  • Endemic mycoses
    ○ Histoplasmosis esp in HIV
    ○ Coccidioidomycosis
    ○ Paracoccidioidomycosis
  • Leishmaniasis
    ○ Generally cutaneous, wet ulcers, non-healing wet ulcers

Other
- Carrion’s disease (Oroya fever) -> Bartonella
- Bolivian haemorrhagic fever
- Oropouche

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
442
Q

Sanctuary sites for Ebola?

A

Testes and Eye

Men can transmit in semen months after being sick, whilst being asymptomatic -> can prompt new outbreak

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
443
Q

You work up a patient for TB but then they tell you they are from Arizona. What should you be considering?

A

Valley fever eg Coccidioidomycosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
444
Q

A 37-year-old Panamanian male presented to hospital in Panama City with an illness of four months duration, characterised initially by weight loss and subsequently by progressive dysphagia, odynophagia, and new oral lesions. At presentation he was unable to swallow fluids and was severely fatigued. There was no history of fever, and his past medical history was unremarkable. A coffee farmer by trade, he lived in the highlands in the west of the country on the family plantation. Aside from the journey to Panama City for treatment, there was no history of travel. On examination, he was cachectic, pale and had non-tender hepatosplenomegaly without lymphadenopathy. An oral examination revealed erythematous ulcerative lesions on his hard and soft palate, oral candidiasis, and poor dentition. Physiological observations were within the normal range, except for a mild tachycardia of 105 bpm. Initial blood tests showed a pancytopenia, with a haemoglobin of 118g/L, white cell count of 1.5x109/L, neutrophil count of 0.5x109/L, and platelets of 88x109/L. Urea and electrolytes, liver function tests and lactate dehydrogenase were all within the normal range. A chest radiograph revealed a large right-sided shadow in the para-mediastinal area. What are your differentials?

A

Histoplasmosis
Leishmaniasis
Actinomycosis
TB
Lymphoma
Paracoccidioidomycosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
445
Q

A 37-year-old Panamanian male presented to hospital in Panama City with an illness of four months duration, characterised initially by weight loss and subsequently by progressive dysphagia, odynophagia, and new oral lesions. At presentation he was unable to swallow fluids and was severely fatigued. There was no history of fever, and his past medical history was unremarkable. A coffee farmer by trade, he lived in the highlands in the west of the country on the family plantation. Aside from the journey to Panama City for treatment, there was no history of travel. On examination, he was cachectic, pale and had non-tender hepatosplenomegaly without lymphadenopathy. An oral examination revealed erythematous ulcerative lesions on his hard and soft palate, oral candidiasis, and poor dentition. Physiological observations were within the normal range, except for a mild tachycardia of 105 bpm. Initial blood tests showed a pancytopenia, with a haemoglobin of 118g/L, white cell count of 1.5x109/L, neutrophil count of 0.5x109/L, and platelets of 88x109/L. Urea and electrolytes, liver function tests and lactate dehydrogenase were all within the normal range. A chest radiograph revealed a large right-sided shadow in the para-mediastinal area. Grocott staining showed budding yeast-like forms. What is the likely diagnosis?

A

Paracoccidioidomycosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
446
Q

Where is paracoccidioidomycosis found and how does it present?

A

Mexico in north to northern regions of Argentina -> think poor male farm workers

Typically asymptomatic pulmonary infection with lymphatic spread -> subsequent immune control leads to granuloma formation -> clearance

In children/young adults -> can get disseminated disease via reticuloendothelial dissemination

80/90% who get disease in adulthood -> reactivation of latent foci months-years after initial infection

Looks like TB!

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
447
Q

How does chronic paracoccidioidomycosis present?

A

Pulmonary infiltrates (77%) and upper respiratory mucosal lesions (>50%).

In the upper respiratory tract, the mouth is most commonly affected, with lesions typically appearing as painful ulcers with ragged borders and areas of haemorrhage.

Involvement of the skin, lymph nodes, CNS and adrenal glands are also well-described but any organ system can be involved and occult disease is common. Disease is multifocal in 75% of cases.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
448
Q

Most sensitive test for paracoccidioidomycoses?

A

Histopathological tissue examination with Grocott or periodic acid-Schiff stains is highly sensitive (>95%), but more invasive. The size and multiple-budding appearance of the Paracoccidioides yeasts is distinguishing from other endemic mycoses, with ‘ships wheel’ or ‘Mickey Mouse’ structures considered pathognomonic.

Culture => can take a month and is affected by site and burden of infection

Serology -> risk of cross-activity with other endemic mycosis, sensitivity of around 80%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
449
Q

Treatment of paracoccidioidomycosis?

A

Mild/moderate: Itraconazole

Severe/CNS: Amphotericin B induction therapy then itraconazole

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
450
Q

What is Coccidioidomycosis also known as? How does it present?

A

Valley fever -> lots in Latin America but can be in Arizona or surrounding states

Symptoms:
- Fatigue
- Fever + night sweats
- Weight loss
- Cough, CP, dyspnoea
- Haemoptysis
- Headache
- Arthralgia
- Looks like TB
Clinical
- Erythema nodosum (25% of pulmonary)
- Erythema multiforme

Can also have dissemianted form with extensive cutaneous involvement and multisystem involvement

CXR
- Pulmonary infiltrates
- Hilar adenopathy
Pleural effusions -> can mimic TB

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
451
Q

Clinical presentation and vectors of Oropouche virus?

A

Symptoms:
- Incubation 3-10 days
- High fever + headache + myalgia/arthralgia + rash ~25%
- Severe: neurological eg meningoencephalitis, haemorrhagic eg gingival bleeding, epistaxis, petechiae
- ?Biphasic -> ~60% have relapse type picture and get sick again
- Low case fatality rate

Ddx:
- Arboviruses eg dengue, Chik, Zika

Vectors: Culicoides paraensis = biting midges + Culex quinquefasciatus, Coquillettidia venezuelensis, and Aedes serratus mosquitoes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
452
Q

What is Culicoides paraensis? What is it a vector for?

A

Oropouche virus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
453
Q

Key facts including epi, host, virus and management of Bolivian haemorrhagic fever?

A

Virus: Machupo virus
Animal: Calomys callosus eg large vesper mouse
Epi:
- Range of reservoir and human movement
Treatment: ribavirin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
454
Q

What are the two key types of filovirus? Where are they endemic?

A

Ebola -> west, tropical rainforest
Marburg -> drier forest or savannah in East

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
455
Q

What are the pathophysiological hallmarks of ebola/marburg infection?

What is the clinical presentation?

A

Microvascular instability with capillary leak + impaired haemostasis +/- DIC

Fever/headache/asthenia/hepatosplenomegaly but no lymphadenopathy/possible early maculopapular rash/N&V&D/bleeding eg from gums, conjunctival haemorrhage, rectum

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
456
Q

Likely reservoir for filoviruses?

A

Fruit bats
Human infection likely occurs due to exposure to infected bat excreta or saliva

Miners/forest workers/spelunkers at high risk, esp for Marburg -> have they been into a cave or mine?

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
457
Q

Possible intermediate hosts for filoviruses?

A

Nonhuman primates esp gorillas and chimps and other wild animals
-> humans come into contact with their blood and bodily fluids

Esp through hunting or butchering

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
458
Q

What causes Tungiasis and how does it present? How do you manage it?

A

Female sandflies -> Tunga penetrans -> burrows into epidermis of humans, often in feet to lay eggs

Pale nodule with dark centre that develops into brown/black with crust, slow growing nodules on feet (periungal regions/interdigital), systemically otherwise well. Due to contact with contaminated soil.

Ix:
Look for parasite and eggs in tissue samples and histopathological specimens
Clinical diagnosis

Mx:
- Check they have received tetanus injection/booster
- Remove flea with sterile needle, disinfect

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
459
Q

Biggest factor contributing to reduction in Malaria from 2000s onwards?

A

ITN = insecticide treated nets

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
460
Q

Key challenges to the control/prevention of Malaria?

A

Need to target outdoor stage, ie most interventions currently aimed at within the home which is unrealistic

  • Financing
  • Drug resistance
  • Vector resistance
    ○ Biochemical
    ○ Behaviour
  • Diagnostic resistance
  • Organisational
  • Climate change
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
461
Q

Key vectors of malaria in Africa?

A

Anopheles
- gambiae
- funestus
- arabiensis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
462
Q

Malaria vector control methods related to life cycle stage?

A

Egg Stage:
Larval Source Management (LSM)

Larval Stage:
Larval Source Management (LSM)

Pupal Stage:
Larval Source Management (LSM)

Adult Stage:
Insecticide-Treated Nets (ITNs)
Indoor Residual Spraying (IRS)
Space Spraying
Personal Protection Measures

Feeding Stage:
Personal Protection Measures

Genetic Control:
Genetic approaches (e.g., genetically modified mosquitoes)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
463
Q

What are the key threats to vector control in Malaria?

A
  1. Insecticide resistance
  2. Invasive vectors eg Anopheles stephensi
  3. Climate change
  4. Diminished access, use and durability of insecticide treated nets (ITNs
  5. Funding
  6. Outdoor transmission (residual transmission)
    • Increase in vectors biting outdoors -> reducing efficiency of indoor vector control tools like ITNs and IRS
  7. Natural disasters, conflict situations and demographic changes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
464
Q

Impact of climate change on malaria vector control?

A

· Warmer temperatures can alter vector survival, abundance and increase the geographical spread of malaria vectors

· Extensive rainfalls and floods can create more breeding sites extending malaria transmission seasons.

· Changes in weather can alter mosquito composition, resulting in higher densities of more efficient vectors.

· Weather changes can affect human behaviour facilitating habits that expose them to vectors e.g sleeping outside in hot weather

· Temperature changes accelerate changes in vector behaviour, enabling them to escape traditional vector control interventions

· Temperature and humidity changes can affect the efficacy of some vector control products

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
465
Q

Why is Anopheles stephensi a problem and what are strategies to address it?

A
  • Historically Asian vector eg India, Iran, Pakistan and Arabian penninsula
  • Efficient vector of urban malaria (plasmodium falciparum and vivax)
  • Breeds in domestic and other artificial water containers, man made pits etc -> urban settings -> current management not build to handle this
  • Can resist high temperatures so can sustain transmission in dry seasons
  • Vector difficult to target with standard vector control

What can be done?
* Increased vector surveillance
* Increase collaboration between different sectors
* Bye laws on water storage especially in urban settings
* Increase vector control strategies that can target the vector e.g larviciding
* On-going coordination meetings led by WHO

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
466
Q

New vector control methods being considered/trialled for malaria vector control?

A
  • Bait stations (attractive targeted sugar
    baits)
  • Spatial repellents
  • Systemic insecticides and endectocides
  • Genetic manipulation, including gene
    drive and self-limiting systems
  • Topical repellents
  • Insecticide treated clothing
  • Housing modification
  • Lethal house lures (EaveTubes)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
467
Q

What are the two classes of Next generation ITNs and IRS products?

A
  1. Dual ITNs (Pyrethroids + new Insecticide)
  2. New IRS eg Pirimiphos-methyl, Clothianidin, eg Broflaniline WP ( Vectron T500, under evaluation)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
468
Q

Definition of chemo prevention wrt Malaria?

A

Administration of a full therapeutic courses of antimalarial treatment to a population at risk, whether infected or not, at defined time points separated by periods without drug exposure people living in endemic areas

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
469
Q

Definition of chemoprophylaxis wrt Malaria?

A

Administration of an anti-malarial drug to an at risk population in such a way as to achieve protective blood levels over the whole of the period of risk sub-therapeutic doses primarily used by non-immune travelers to malaria endemic areas

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
470
Q

What are the WHO 5 recommendations for Malaria chemoprevention in endemic areas?

A
  1. Intermittent preventive treatment of malaria in pregnancy (IPTp)
    - give from 13 weeks gestation onwards, at least 3 doses during pregnancy, give at antenatal visits, allow 1 month between treatment times
  2. Perennial malaria chemoprevention (PMC), previously known as intermittent preventive treatment in infants (IPTi)
    - From 10 weeks of age onwards up to 24 months
    - Sulfadoxine-Pyrimethamine normally used, with ACT now being considered
  3. Seasonal malaria chemoprevention (SMC) previously known as intermittent preventive treatment in children (IPTc)
    - Door to door/community based intervention
    - Use Sulfadoxine-Pyrimethamine and Artesunate Combination Therapy (SPAQ)
  4. Intermittent preventive treatment in school aged children (IPTsc)
  5. Post-discharge malaria chemoprevention (PDMC)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
471
Q

What does SPAQ stand for in the context of malaria chemoprevention?

A

Sulfadoxine-Pyrimethamine and Artesunate Combination Therapy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
472
Q

What are the two vaccines approved by WHO for malaria?

A
  1. RTS,S/ASO1E (Mosquirix ®): Oct 2021
  2. R21 /Matrix M : Sep 2023
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
473
Q

Target of malaria vaccines?

A

Circumsporozoite protein

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
474
Q

Two Malaria trials and findings

A

RTS,S/ASO1E (Mosquirix ®)
- 3 primary vaccinations were given prior to the season followed by an annual booster dose until children reached 5 years of age
- Compared with and to seasonal chemoprevention
- RCT showed non-inferior to seasonal chemoprevention alone, even higher efficacy when combined
- Reduction in incidence, hospital admissions and deaths

R21 /Matrix M
- RCT
- Reduction in time to first episode and all episodes, seasonal > standard by ~5%
- Efficacy held at all and seasonal sites at 24 months

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
475
Q

With respect to Chemoprevention of malaria, what do IPTp and SMC stand for?

A

Intermittent preventative treatment in pregnancy
Seasonal malaria chemoprevention

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
476
Q

Mosquito genuses that transmit Lymphatic filariasis?

A

Anopheles
Culex
Aedes
Mansonia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
477
Q

Mosquito genus for Japanese encephalitis?

A

Culex (tritaeniorhynchus)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
478
Q

Mosquito genus for West Nile?

A

Culex (pipiens)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
479
Q

Mosquito genus for Yellow fever?

A

Aedes, Haemagogus, Sabethes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
480
Q

What does Aedes transmit?

A

Dengue
Zika
Chikungunya
Lymphatic filariasis
Yellow fever

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
481
Q

What is the mosquito life cycle?

A
  • Adult female mosquito mates soon after emergence from pupa and stores spermathecae (the sperm from male mosquitoes) to fertilise batches of eggs
  • After the female mosquito blood feeds, egg development starts
  • Female searches for appropriate quality of water for egg laying
  • Different species of mosquito make different choices as where to lay their eggs
  • Larvae may hatch soon after eggs are laid or after some time depending on conditions
  • Larvae swim, feed, breathe air and develop through 4 larval stages called instarts
  • 4th instar metamorphoses into pupa
  • Pupa swims and breathes are but do not feed
  • Pupa metamorphoses into adult mosquito which emerges at the water surface, stretches its wings and flies away
  • Both sexes of adult obtain energy from sugary plant juices

-Only the adult female feeds on blood to get proteins to feed her young

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
482
Q

Terms used to describe whether mosquito enters house or not to feed?

A

Endophagic
Exophagic

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
483
Q

Terms used to describe whether mosquito like to digest inside or outside?

A

Endophilic or exophilic

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
484
Q

Aedes aegypti properties in terms of
- entering house or not to feed
- digesting inside or outside
- who it likes to bite

A

anthropophilic, exophagic and exophilic

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
485
Q

Anopheles gambiensae

A

anthropophagic, endophagic, endophilic

bites late at night after people are asleep

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
486
Q

Definition of a random sample

A

Each individual/each household etc has equal probability of being included

Can use random cluster sample if large population and homogenous distribution of disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
487
Q

Definition of prevalence

A
  • Number of cases of disease in a defined population at a given point in time
  • Is affected by duration
    o Ie if you increase duration of illness then it can drive up prevalence because you capture more people with the illness at that time of disease, but incidence is unaffected
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
488
Q

Definition of incidence

A
  • The number of new cases of disease in a defined population ‘those at risk’ over a specified period of time
    o Population at risk should not only be those people but how much time they spend at risk
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
489
Q

Definition of incidence risk

A
  • Number of new cases that arise in population and divides them by the number of people who started off at risk
  • Counts all people at start, then at end of period of observation tells you how many people got ill during that period
    o Irrespective of whether they were all at risk at that period of time
    o Easier to calculate than incidence rate, as you just count how many people you start with and how many have developed outcome disease at the end
    o But becomes inaccurate if you lose people to follow up as you can’t verify them so your numerator lacks accuracy

Defined as: number of new cases in specified period/population of those disease free at start of period of observation
If they are not within the numerator then they can’t be within your denominator

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
490
Q

Definition of incidence rate

A

Number of new cases that arise in population in specified period/sum of length of time during which each person in the population is at risk

  • Denominator: person-time at risk
  • So take every individuals person time at risk over given period of time and add them all together
  • If you lose people to follow up, the numerator will still lack accuracy
    If a disease only gives you partial immunity eg Covid, then you can be part of the numerator again when you become at risk again
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
491
Q

Advantages and disadvantages of case reports?

A

Adv: rapid sharing of new info about rare occurrences and useful in hypothesis generation

Dis: one experience, ?coincidental, lack of comparison group

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
492
Q

Advantages and disadvantages of cross sectional surveys?

A

o A representative sample of a population is studied to quantify a disease, an exposure or both
o ‘Snapshot’ of population at a single point in time
o Exposure and disease assessed simultaneously
§ Eg surveys
§ Trends over time can be examined using repeated cross-sectional surveys eg polling before elections

Adv: quick and relatively easy, can measure prevalence, can measure prevalence of RF, can measure demand for service (for health service planning), hypothesis generation

Dis: prevalence subject to singular point in time, can’t measure incidence, can’t determine causality

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
493
Q

Advantages and disadvantages of case control studies?

A

o People with disease (case) and people without the disease (control) are compared by looking at past exposure of each group to potential causes
§ Retrospective

o Adv: Can be useful for investigating causation, particularly if disease is rare or long ‘incubation’ period after exposure (when cohort studies impractical), quick and inexpensive, can examine multiple exposures

o Dis: selection bias (ensuring case population is representative of larger control population apart from outcome is hard), impractical if exposure is rare, not always certain that exposure preceded disease, cannot measure disease incidence

OR: yes/no for cases compared to yes/no for controls for given exposure

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
494
Q

Advantages and disadvantages of cohort studies?

A

o Study people with a risk factor to see if they develop particular diseases
o Can do descriptive or analytic cohort studies
o Recruit people free from disease, classify participants at baseline as exposed or unexposed, follow both to determine outcome, compare incidence of outcome between exposed and unexposed
o Usually prospective

o Adv: can measure incidence, useful for prognosis and causation, useful for rare exposures, can examine multiple outcomes, can show exposure comes from disease, can measure exposure and outcome data directly so more accurate and less risk of bias

o Dis: expensive & time consuming, impractical/efficient if outcome is rare, vulnerable to losses to follow up

o Retrospective cohort
§ Construct cohort based on past records
§ Efficient if accurate outcome data
§ Problematic if poor data on outcome

Nested case-control
At end of cohort study when outcomes have occurred, you can select cases (participants who developed outcome of interest during follow-up) and controls (participants who did not develop outcome during follow-up)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
495
Q

Advantages and disadvantages of RCTs?

A

o Pre-specify an outcome
o Investigator allocates exposure
o Randomisation: enables two groups to be balanced for factors/characterisations which could influence the outcome, so you isolate the effect of only the intervention you want (treatment or prevention)

o Adv: strongest evidence of effect of intervention, can examine more than one outcome, prospective, eradicates effect of bias

o Dis: expensive, time consuming, if not properly planned then can be too small or too short, not always practical (ie if no. of patients needed is prohibitively high, if randomisation is unethical), trials do not necessarily reflect what happens when intervention is delivered routinely ie in real life

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
496
Q

Infections that can give you a hepatitis

A

Hepatitis ABCE
Leptospirosis
Arboviruses: Yellow fever virus (AST>ALT)
VHF: Lassa fever
Herpes viruses: CMV, EBV
Malaria

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
497
Q

Key facts Hepatitis A

A

Faecal oral transmission, incubation 4-8 weeks

  • Children esp <6 ~70% asymptomatic, but spreaders, >6 70% jaundiced
  • Adults: jaundice, fulminant hepatitis +/- liver necrosis, risk of failure if pre-existing liver disease
    • If symptoms then can be symptomatic for months
    • Spontaneous resolution over 1-2 weeks
    • No chronic HAV

Diagnosis: serology IgM/IgG

Vaccine
○ Effective in 95% cases, immunity for 20 years
Can’t give to kids <6 months -> need anti-hepA immunoglobulin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
498
Q

Key facts Hep C

A

Transmission: blood borne, bodily fluids

Incubation: 2 weeks - 6 months

Who:
- (IVDUs/haemophiliacs issue/needles/tattoos/dialysis)
- 15-25% spontaneous clear
- 75-85% chronic infection

S/S:
- <30% symptomatic
- Liver failure 1/2%

Diagnosis: hep C total Ab +/- Hep C RNA
- Ab can persist for whole life
- If immunocompromised or high risk (ie might not mount full immune response or slow one) then just do hep C RNA directly
- Some Ag tests available

Mx: DAAs upon diagnosis (based on detectable HCV RNA)

Vaccine: none, reinfection common esp in high risk groups

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
499
Q

Two types of outbreaks for Hepatitis E?

A
  • Sporadic acute jaundice
    ○ Genotypes 3/4
    ○ Consumption of undercooked or raw pork/game meat and shellfish
    ○ High income countries
  • Acute jaundice in Africa and Asia
    ○ Genotypes 1/2
    ○ Faecal contamination of drinking water
    ○ Humanitarian crises
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
500
Q

In low income countries most people will be exposed to what hepatitis virus?

A

Hepatitis A, so most people will have Abs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
501
Q

How should you treat a baby if pregnant mum is Hep B positive?

A
  • Treat child with tenofovir or TAF from week 30 to week 4 post-partum
  • HBV vaccine and HBIG on first day of life
    HBIG not really available outside high income countries
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
502
Q

Hepatitis B core Ag (HbcAg) generally means

A

Acute infection, if persists >6 months then = chronic infection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
503
Q

Life cycle of Entamoeba histolytica

A

Ingest cyst
Swallow cysts
Pass through stomach as resistance to acid
Exist/hatch in small intestine
Release of trophozoite
Multiplication
Cysts form
Hatching in stool -> infect others

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
504
Q

What index can help predict cirrhosis in HbsAg +ve?

A

AST to Platelet Ratio Index

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
505
Q

Which demographic does E.histolytica liver disease most affect?

A

Adult males
no sex differences in Amoebic dysentry

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
506
Q

Diagnostic methods for Entamoeba histolytica?

A

· Wet-film microscopy (but note cannot distinguish between E. histolytica and E. dispar)
○ Concentrated, fixed and stained faecal samples
· Serology - Relatively cheap. Usually strongly positive in ALA, can persist
· Tissue biopsies - likely histolytica if seen in tissue
· Antigen detection – Not all distinguish Eh/Ed. Reliable?
· Nucleic acid methods – Real-time PCR, LAMP, metagenome
· Diagnosis of ALA – Imaging, symptoms

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
507
Q

Treatment for E.histolytica

A
  • Invasive amoebiasis is almost exclusively treated with metronidazole or other 5-nitroimidazole drugs (equally but no more effective)
  • Should be followed by a luminal amoebicide– choice depends on what is licensed locally
  • Paromomycin is most effective for luminal E. histolytica
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
508
Q

Where do Giardia cysts colonise?

A

Small intestine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
509
Q

Where do Entamoeba cysts colonise?

A

Large intestine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
510
Q

Possible pathophysiological mechanisms of Giardia?

A
  • Malabsorption of glucose, water, Na+
  • Diffuse microvillus shortening
  • Loss of epithelial barrier function -> increased intestinal permeability
  • Apoptosis of enterocytes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
511
Q

How do you diagnose Giardia?

A
  1. Wet film microscopy of faecal samples
    ○ Cysts easy to recognise, so specificity is high but sensitivity low
  2. Direct fluorescence Ab staining of cysts
  3. Ag detection (ELISA/card; slightly less sensitive than DFA)
  4. Conventional, multiplex & real time PCR
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
512
Q

How is Giardia transmitted? What permits its transmission/issues with it?

A
  • Food, water, person to person
  • Infectious dose is very small ~10 cysts
  • Cysts are resistant to chlorine and other disinfectants
  • Efficacy of conventional wastewater treatment processes at removing cysts is limiting
  • Cysts remain viable for weeks in cold water
  • Animal reservoirs of some genotypes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
513
Q

Treatment of Giardia

A

Metronidazole (1, avoid in first trimester) / Tinidazole (seems to be slightly better) / Nitazoxanide

Paromomycin appears to be effective and is regarded as safe in pregnancy

Treatment failures with nitroimidazoles are not uncommon. Drug resistance is a growing problem.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
514
Q

What three species of trichomonads affect humans?

A
  1. Trichomonas vaginalis
  2. Trichomonas tenax
  3. Pentatrichomonas hominis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
515
Q

How to diagnose trichomonas vaginalis?

A
  • Still largely by wet-film examination
  • Culture will detect 10-30% more but takes longer
  • Rapid antigen detection kits provide the speed of wet film & better sensitivity than culture
  • PCR and other nucleic acid amplification tests (NATS) probably the most sensitive but not yet in routine use
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
516
Q

How is trichomonas vaginalis transmitted? Why is it an issue?

A

Sexually transmitted
Linked to low birthweight and also higher risk of HIV acquisition and transmission

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
517
Q

Clinical manifestations of trichomonas vaginalis?

A

Most common signs and symptoms in women:*
- Purulent vaginal discharge
- Vaginal and vulvar erythema
- Urethra commonly infected causing dysuria

In men:
- Urethral discharge and/or dysuria
- Spontaneous loss of infection seems to be quite frequent

The overwhelming majority of infections are asymptomatic (98.5% in men, 73.3% in women)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
518
Q

Treatment of trichomonas vaginalis?

A
  1. Single 2g dose of metronidazole (MTZ)
  2. Tinidazole -> sig more effective but 10x more expensive (also avoid in pregnancy)
  3. The most recent CDC STI treatment guidelines now recommend metronidazole 500mg PO BD (women); single 2g dose (men)
    Metronidazole resistance is a rare but real problem
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
519
Q

Key features of Dientamoeba fragilis?

A
  • Amoeboid appearance but is related to trichomonads
  • Pathogenicity is controversial and certainly variable
  • Reported symptoms are non-specific (e.g. diarrhoea, abdominal pain)
  • The trophozoite will not be found by light microscopy unless specimens are rapidly fixed and stained
  • If use PCR, infection rate is high in the general population

Treatment:
- Metronidazole (not particularly effective)
- Clioquinol - more effective in reliving symptoms
- Tetracycline - also used
- Secnidazole - may be the best

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
520
Q

What is the only ciliate to infect humans and relevant features?

A

Balantioides coli

  • Both cyst and trophozoite are much larger (> 50 μm) than other gut protozoa
  • Very common in pigs, but rare in humans
  • A large bowel parasite
  • Frequently asymptomatic but can occasionally invade like E. histolytica and cause colitis, dysentery and death
  • Metronidazole seems to be effective but few recent reports available
  • Cysts and nucleus are huge
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
521
Q

Definition of XDR TB?

A

Resistance to

  1. Isoniazid (INH) and Rifampicin (RIF), the two most powerful first-line TB drugs (this is what defines multidrug-resistant TB, or MDR-TB).
  2. Fluoroquinolones, a class of antibiotics that are key second-line drugs.
  3. At least one of the injectable second-line drugs, such as amikacin, kanamycin, or capreomycin.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
522
Q

Definition of pre-XDR TB?

A
  • Rif resistance +/- isoniazid resistance + resistance to fluoroquinolone (mof/levofloxacin)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
523
Q

Definition of MDR TB?

A
  • Combo of isoniazid and rifampicin resistance
  • Unusual to have Rif resistance without isoniazid resistance
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
524
Q

What is DOT for TB?

A

Key Goals of DOT:
Ensure Adherence
Prevent Drug Resistance
Monitor Side Effects:
Support Patients

Types of DOT:
In-person DOT: Patients visit a clinic or a healthcare worker comes to the patient’s home to observe them taking the medication.

Electronic DOT (eDOT): Patients record themselves taking their medication via video, which is reviewed by healthcare workers remotely. This method is increasingly being used where in-person observation is not feasible.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
525
Q

What are “BPaL” and “BPaLM” for TB?

A

Used for MDR and XDR, treat for 6 months (prior regimens used 9-18 months)
Only for adults, no CNS/disseminted/

MDR you can use BPaLM:
bedaquiline (B), pretomanid (Pa), and linezolid (L) + moxifloxacin (M)

BPaL:
bedaquiline (B), pretomanid (Pa), and linezolid (L)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
526
Q

SEs linezolid

A

Drop FBC
Eye toxicity
Peripheral neuropathy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
527
Q

SEs bedaquiline

A

Prolong QTc -> risk of sudden death
Hepatitis
N&V&Arthralgia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
528
Q

SEs pretomanid

A

Very expensive

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
529
Q

Who can you use a urinary LAM in?

A

Only use if suspecting TB in HIV
Only approved for patients with low CD4 count
Only looks for active disease, ie not passive

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
530
Q

Who do you blanket give TB preventative therapy to and what do you rule out in them?

A

Rule out active disease

Give preventative treatment to children under 5 and HIV positive individuals/high risk

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
531
Q

If person has latent TB and has had contact with MDR person?

A

Empirical 6 months levofloxacin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
532
Q

Ddx for slow growing small facial lesion that has ulcerated in tropical setting. Itchy but not painful with no lymphadenopathy and systemically well.

A

Cutaneous leishmaniasis - localised painless ulcer with raised edges
Sporotrichosis -> Sporothrix schienckii
Cutaneous TB
Balamuthia mandrillaris (amoeba)
Francisella tularensis (Tularaemia) -> northern hemisphere only
Cutaneous anthrax - but will appear dark

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
533
Q

Why is Amastigotes also called the Leishmanial stage?

A

Its the form leishmania takes in the vertebral host
otherwise = protist cell that does not have visible external flagella or cilia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
534
Q

What causes granulomatous amoebic encephalitis?

A

Balamuthia mandrillaris

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
535
Q

What causes Sporotrichosis?

A

Sporothrix schenckii - dimorphic fungi
Found in soil/on plants
Enters skin through direct inoculation eg thorny plants/animal scratch
Papule -> tender ulcer
Lesions can spread along draining lymphatic vessels

?farmer/gardener
Hyperendemic in parts of Andes -> children, facial lesions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
536
Q

Key facts about cutaneous leishmaniasis?

A

Protozoa = genus Leishmania
Anthroponotic or zoonotic (reservoir small animals)
Transmission: sandflies
- Lutzomyia (New World)
- Phlebotomus (Old World)

Incubation period: weeks to months

Clinical: depends, most common is localised

Old world (Africa/Asia/Europe): >50% heal spontaneously
New world (Latin America): <20% heal sponteanously

Treatment:
OW/NW (but not L.Viannia-complex): intralesional pentavalent antimonial eg sodium stibogluconate SSG, paramomycin cream +/- cryotherapy or thermotherapy

Systemic tx: Pentavalent Antimonials (e.g., Sodium Stibogluconate or Meglumine Antimoniate) injections 20-28days OR Miltefosine OR Azoles
2nd LINE: Amphotericin B OR ambisome

L.guyanensis: pentamidine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
537
Q

Options for diagnosis of cutaneous leishmaniasis?

A
  1. Skin scrapping and Giemsa staining -> amastigotes
  2. Culture on FNA/biopsy samples
  3. Montenegro or Leishmania skin test -> test for delayed immune response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
538
Q

Causes of ptosis?

A

Oculomotor damage -> innervates the levator palpebrae superioris
- Lesions can result in complete ptosis

Sympathetic damage eg Horner’s -> partial ptosis via superior tarsal muscle

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
539
Q

Causes and features of bacterial meningitis?

A

Streptococcus pneumonia
Neisseria meningitides
Streptococcus suis (Asia)

Rapid onset, high fever, meningism, CN involvement less common

CSF: cloudy, high leucocyte count, polymorphs, low glucose

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
540
Q

Causes and features of TBM?

A

Mycobacterium TB

Hx of several days of illness, slightly less abrupt, CN involvement common

CSF: clear, high CSF protein and low glucose

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
541
Q

Causes and features of Cryptococcal meningitis?

A

Cryptococcus neoformans

Hx: subacute onset, severe headache, CN involvement

CSF: normal in at least 25% of cases

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
542
Q

What is the relevance in Hep B of
- HbsAg
- HbcAg
- HbCAb
- HBV IgM anti-Hbc

A
  • Hepatitis surface Ag (HbsAg) to have hep B, >6 months = chronic
  • Hepatitis B core Ag (HbcAg) generally means acute infection
  • Isolated hep B total core Ab (HbCAb) can mean prior infection
  • Core IgM Ab = acute infection
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
543
Q

Key Hepatitis B facts

A

Transmission: body fluids, blood etc

Incubation: 1-6 months

Who:
* Acquiring as child -> MUCH higher risk of CHRONIC infection, worse if vertical transmission
○ <1 year: usually asymptomatic
○ >1 year: symptomatic infections
* 30% symptomatic, 95% adult cases will resolve, 1% liver failure
· 5-10% of adults get chronic infection
· Only need a small amount of exposure to blood with high viral load

Diagnosis:
- Hepatitis surface Ag (HbsAg) to have hep B, >6 months = chronic
- Hepatitis B core Ag (HbcAg) generally means acute infection
- Serology for HBV Core IgM

Vaccine:
Recombinant HbsAg - 3 doses - 95% effective in healthy people

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
544
Q

Key facts Hepatitis E

A

Transmission: Faecal-oral, domestic pigs/deers

Incubation: 4-8 weeks, often preceded by prodrome and diarrhoeal illness

Who:
- Sporadic acute jaundice
○ Genotypes 3/4
○ Consumption of undercooked or raw pork/game meat and shellfish
○ High income countries

  • Outbreaks of acute jaundice in Africa and Asia
    ○ Genotypes 1/2
    ○ Faecal contamination of drinking water
    ○ Humanitarian crises
  • Mortality in pregnant women can be up to ~20%
    S/S:
  • Mild or symptomatic
  • Typically resolves spontaneously over 1-2 weeks
  • Liver failure rare except in pregnancy (acute liver failure in 20%), chronicity recognised in immunocompromised

Diagnosis:
- Serology (IgM/IgG) and molecular testing (HEV RNA)

Vaccine:
Licensed in China, global trials ongoing in women of child bearing age

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
545
Q

Options for treatment of acute Hepatitis B infection?

A
  • RCTs have not demonstrated efficacy but are safe
  • Lamivudine/entecavir/tenofovir if 2 of
    ○ Hepatic encephalopathy
    ○ Bilirubin >10mg/dL (171micromol/L)
    ○ INR>1.6
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
546
Q

What does HBV/HCV co-infection put you at risk of?

A
  • 7.4% globally
  • Much more likely to develop chronic HBV 30%
  • HIV/HBV coinfection indication for treatment with ART against HBV (tenofovir/emtricitabine or lamivudine backbone)
  • Without treatment -> progression to cirrhosis can be rapid
  • Treatment does not totally stop disease progression
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
547
Q

How does chronic HBV infection lead to HCC?

A
  1. Acute inflammation -> carcinogenic properties of virus directly -> HCC
    ○ Uncontrolled hep B can cause liver cancer due to integration into host genome
  2. Acute inflammation -> Fibrosis -> cirrhosis 20% (30% decompensated) -> HCC 1-4% annually
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
548
Q

What are the problems with treating chronic HBV? What approach should be taken?

A

Nucleot(s)ide reverse transcriptase inhibitors (NRTI) suppress viraemia eg TDF
But CANNOT get rid of DNA in nucleus of hepatocytes due to cccDNA forming minichromosome that remains despite therapy
Means virus can reactivate later in life eg if immunosuppressed

  • Treat
    ○ Functional cure -> does not rule out risk
  • Minimise superimposed risks
    ○ No etoh
    ○ Lower infection risk
    ○ Control metabolic syndrome
    ○ Behaviour -> eg avoid groundnuts, cooking equipment with led etc
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
549
Q

What does elimination of a disease mean?

A
  • Reduction to zero of the incidence of a disease in a defined geographical area
  • Continued interventions required to present re-introduction
    Eg Polio in the Americas
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
550
Q

What does eradication of a disease mean?

A
  • Reduction to zero of the incidence of a disease in a defined geographical area
  • Continued interventions required to present re-introduction
    Eg Polio in the Americas
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
551
Q

What are some requirements for elimination or eradication of a disease?

A
  • An effective intervention is available to interrupt transmission
  • Surveillance: Practical diagnostic tools of sufficient sensitivity and specificity to detect levels of infection that can lead to transmission are widely available
  • No animal reservoir
  • The disease must be widely recognised to be of public health importance
  • Elimination must be perceived as a worthy goal by all levels of society
  • A technically feasible intervention must have been field-tested and found effective
  • Political commitment must be obtained at the highest levels (WHA resolution)
  • Advocacy plan must have been prepared
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
552
Q

What did the London declaration on NTDs 2012?

A

Resolved to sustain, expand and extend programmes that ensure the necessary supply of drugs and other interventions to help eradicate Guinea worm disease, and help eliminate by 2020 lymphatic filariasis, leprosy, human African trypanosomiasis and blinding trachoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
553
Q

NTDs targeted for global eradication by WHO roadmap of 2012?

A
  • Yaws
  • Guinea worm
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
554
Q

NTDs targeted for global elimination by WHO roadmap of 2012?

A
  • Onchocerciasis in South America
  • Human African trypanosomiasis due to T.b.gambiense
  • Schistosomiasis
    – in China, Eastern Mediterranean, Caribbean, Indonesia, Mekong basin
    – In the Americas and Western Pacific Region
  • Chagas disease through blood transfusion (2015); intra-domiciliary
  • Rabies in SE Asia and Western Pacific
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
555
Q

NTDs targeted for elimination as a global health problem by WHO roadmap of 2012?

A
  • Blinding Trachoma
  • Leprosy
  • Visceral leishmaniasis in the Indian sub-continent
  • Lymphatic filariasis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
556
Q

What are the challenges with tests to certify elimination and for subsequent surveillance?

A
  • Large numbers of people, vectors or intermediate hosts may need to be sampled
    ○ Tests should be high throughput
    ○ Pooling can save time and money
    ○ Tests do not necessarily need to be at the point of care
  • Sensitivity should be high
  • False positives must be kept to a minimum
  • Tests must be highly specific
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
557
Q

Intervention for lymphatic filariasis to eliminate as NTD?

A

○ Annual or twice yearly MDA with single dose ivermectin and albendazole, plus DEC in regions where there is no onchocerciasis
– reduces microfilaraemia but does not kill adult worms

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
558
Q

Proposed new leprosy targets for 2030?

A
  • Zero Grade 2 disability (G2D) among paediatric leprosy patients
  • Reduction of new leprosy cases with G2D to less than one case per million population
  • Confirmed absence of children with leprosy for 5 consecutive years
  • Zero countries with legislation allowing discrimination on basis of leprosy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
559
Q

Key differences between Human African Trypanosomiasis causes?

A

T. brucei gambiense
- Insidious onset (“sleeping sickness”)
- No confirmed animal reservoir
- POC screening test available (lateral flow HAT Sero-K-SeT)
T. brucei rhodesiense
- Acute onset - febrile illness
- Zoonosis
- Blood film usually positive

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
560
Q

Treatment for T. gambiense?

A

– Until 2019
* Stage 1: Pentamidine IM
* Stage 2: Nifurtimox + eflornithine IV

– Now
* Fexinidazole is an effective oral treatment for stage 2 T. gambiense

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
561
Q

Treatment for T.b. rhodesiense?

A

○ Stage 1: Suramin IV (stage 1)
○ Stage 2: Melarsoprol IV + prednisone (stage 2)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
562
Q

Proposed overall goal by 2030 for NTD SDG?

A
  • 90% reduction in the number of people requiring interventions against NTDs
  • 100% of population at risk protected from out of pocket health payments due to NTDs
  • 100 countries have eliminated at least 1 NTD
  • 2 NTDs have been eradicated
  • DALYs lost to NTDs reduced by 75%
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
563
Q

How is trachoma transmitted?

A
  1. Flies
  2. Fomites
  3. Fingers
    Within Family or among close Friends

5Fs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
564
Q

Treatment of trachoma? old vs new

A
  • Initially tetracycline ointment BD for 6 weeks
  • Trial by Prof Bailey RCT, single-blinded in 2 Gambian villages showed single dose PO Azithromycin (20mg/kg) showed better adherence and treatments were equivalent
    Reinfection was common

Now also
- Surgery
- Facial cleanliness + hygiene

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
565
Q

WHO recommendations for azithromycin for trachoma?

A

Baseline survey:
- 5 years MDA where TF ≥30% in 1-9 year-olds
- 3 years MDA where TF 10-29.9%
- 1 round of MDA where TF 5-9.9%
Impact surveys:
- 1-5 years after SAFE interventions began
Surveillance surveys:
At least two years after an impact survey has shown the TF prevalence to be <5% in 1-9 year olds

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
566
Q

What is Trachoma caused by?

A

Chlamydia trachomatis
- Bacterium
- Intracellular organism (biphasic developmental cycle)
Infects mucosal epithelial cells of genital tract and conjunctiva

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
567
Q

Diagnostic tests for Yaws?

A
  • Serological
    ○ TPPA (treponema pallidum partial agglutination test)
    § Stays positive for life
    ○ RPR/VDRL
    § Titres fall following treatment
    § Less specific - eg low level false positives from malaria
  • Molecular
    PCR can differentiate sub-species
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
568
Q

Basic facts and epi of Yaws?

A

Treponema pallidum pertenue
Found in warm/humid environment
○ West Africa
○ South America
○ Pacific
○ South-East Asia

Children = main reservoir of siease
Transmission
○ Skin to skin contact
○ ?Flies

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
569
Q

Stages of Yaws

A

Primary:
§ Painless ulcer or nodule (>1cm, raised, solid) in trop world known as papillomas
§ More papillomas in West Africa as proportion of cases
§ These particular lesions heal within a few weeks

Secondary
Then spread of bacteria -> general illness, disseminated
§ Destructive lesions of skin/soft tissue
§ Face, nasal mucosa, bones of lower limbs
§ No neuro involvement/no CVS disease

Tertiary
§ Asymptomatic between stages
§ Can become latent at any stage
§ No clinical signs of disease
§ Latent cases -> important source of reinfection and onward transmission
§ Ratio latent : clinical between 6-10:1

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
570
Q

What two countries have ?eliminated Yaws?

A

India and Equador

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
571
Q

What rapid tests exist for diagnosing Yaws?

A

Dual Path Platform (DPP) Syphilis Screen and Confirm Test:
- Detects treponemal and non-treponemal Abs
- Can differentiate between an active infection (non-treponemal antibodies present) and a past infection (treponemal antibodies alone)

Treponemal-Specific Rapid Diagnostic Tests: treponemal Abs only so can’t distinguish between active and past infection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
572
Q

What did the YAWS3 trial do/show?

A

YAWS3 trial

One round of MDA, followed by targeted treatment of active cases and their contacts VS Three rounds of MDA at six-month intervals before transitioning to targeted treatment.

○ Three rounds of mass azithromycin administration for yaws eradication
○ Multiple rounds of treatment enable opportunity to address treatment reservoir in addition to acute cases

A single round of MDA significantly reduced the prevalence of yaws, it was insufficient to eliminate transmission in the long term. Reintroductions of yaws occurred due to untreated latent cases or people who missed the MDA. In contrast, the strategy of three rounds of MDA was more effective in maintaining lower prevalence and reducing the risk of re-emergence.

Additionally, the trial highlighted the need to monitor for potential macrolide resistance, as azithromycin-resistant strains of Treponema pertenue had been detected in some cases

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
573
Q

Big issues with Yaws eradication?

A

Poor data on epi + serological same amongst different species of treponema
Limited funding
MDA with azithromycin creating resistance (de-novo mutations)
Potential for animal reservoirs -> similar infections in non-human primates

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
574
Q

Strategy for Yaws eradication?

A

– Mapping to identify current foci
– Mass treatment with
azithromycin (likely with multiple rounds)
– Surveys to identify cases & contacts
– Mop-up Treatment
– Integrate with other skin NTDs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
575
Q

What are the types of endemic trepanematosis?

A

T.pallidum ssp pallidum -> Syphilis
T.pallidum ssp pertenue -> Yaws
T.pallidum ssp endemicum -> Bejel
T.carateum -> Pinta

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
576
Q

Key facts about Pinta?

A
  • T.carateum
  • Found in warm, humid environments
    ○ Restricted to Latin America
  • Active cases in young adults are main reservoir of infection
    Transmission: skin to skin contact
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
577
Q

Key facts about Bejel?

A
  • “Endemic syphilis”
  • T.p.endemicum
    ○ Found in dry environements eg
    § The gulf
    § The sahel
  • Children = main reservoir

Transmission via
○ Skin to skin contact
○ Indirect contact via utensils

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
578
Q

Successes and obstacles of guinea worm eradication?

A

What could help with success?
- Easily identifiable disease
- Relatively simple and vulnerable life-cycle
- Cheap, easy interventions
- Community focused interventions
- Vertical health programme
- Political and economic support

Obstacles to success?
- Insecurity (political etc)
- Population movement
- Donor fatigue
- Community fatigue
- Atypical transmission (dogs get infected by paratenic host)
- Pseudo-presentation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
579
Q

What is Dracunculiasis medinensis?

A

Guinea worm
60cm large nematode

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
580
Q

What is a paratenic host?

A

An organism that harbors a parasite but is not necessary for the parasite’s development to its adult or infective stage. In other words, the parasite can survive and remain infective within the paratenic host, but it does not undergo any further development there. The host simply acts as a temporary “carrier” or “reservoir” for the parasite until it reaches its definitive host, where it will complete its life cycle.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
581
Q

Life cycle of guinea worm rule of 14?

A
  • Lives for 10-14 months as worm in human host
  • 14 days as larvae in water
  • 14 days developing as larvae in vector until it can infect a human again
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
582
Q

What are some skin NTDs?

A
  1. Yaws
  2. Leprosy (Hansen’s disease)
  3. Onchocerciasis
  4. Lymphatic filariasis (lymphoedema and hydrocele)
  5. Buruli ulcer
  6. Cutaneous leishmaniasis
  7. Mycetoma, chromoblastomycosis, and other deep mycoses
    ( including sporotrichosis)
  8. Post-kala-azar dermal leishmaniasis
  9. Scabies
  10. Tungiasis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
583
Q

What causes buruli ulcer and who does it occur in? How is it transmitted?

A

Mycobacterium ulcerans

WHO
- Poor inhabitants, remote rural areas, limited access to healthcare services
- All ages and socioeconomic groups
- Africa ~50% aged <15 years
- Australia: adults mostly affected

TRANSMISSION
- Trauma -> introduction of mycobacterium to skin
- Occurs in people living close to lakes, rivers, marshy areas, mining, deforestation or flooding
- Inoculation into skin by
○ Aquatic insects
○ Mosquitoes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
584
Q

What drives pathology in M.ulcerans?

A

Buruli ulcer toxin called mycolactone

Variants driven by chemical modification of side chain of toxin:
○ A/B: African strains
○ C: Australia
○ D: Asia

Pathogenesis driven by toxin?
○ Low concs of toxin cause suppression of dendritic cells, macrophages, T-cell function abrogating cytokine and chemokine secretion in response to mitogens
Downregulation of inflammatory mediators, lipid metabolism

Early lesions:
- Clumps extracellular mycobacteria
- Subcutaneous fat necrosis
Sparse inflammatory infiltrate

Late lesions:
- Areas of acute and chronic inflammation including:
* few mycobacteria
granulomatous inflammation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
585
Q

Clinical manifestation of buruli ulcer?

A

Incubation: 32-264 days (mean 4.5 months)
No constitutional symptoms associated
Lesions appear on all parts of body but predominantly on the limbs (up to 85%)
Bones involvement (6-25% of lesions in Benin)

Active disease:
- Non-ulcerative (papule, nodule, plaque, oedema)
- Ulcerative forms and osteomyelitis
- Painless, NO LN involvement (usually)
- firm, immobile

If not treated:
Ulceration -> necrosis -> sloughing
- indurated/undermined edges, painless

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
586
Q

Ddx for buruli ulcer?

A
  • Abscess—————————Painful
  • Lipoma———————– Mobile
  • Ganglion————————-Close to joints
  • Tuberculous lymphadenitis—-Constitutional symptoms
  • Onchocerciasis nodule———-Painless, long history
  • Subcutaneous infections such as fungal infection
  • Tropical phagedenic ulcers
  • Arterial and venous insufficiency
  • Diabetic ulcers
  • Sickle cell ulcer
  • Cutaneous leishmaniasis
  • Extensive ulcerative yaws
  • Haemophilus ducreyi ulcers
    Non Buruli ulcers are often painful
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
587
Q

How to take sample to test for buruli ulcer? How to test sample?

A
  1. Swab
    ○ Get under the undermined edge of ulcerative lesions
  2. FNA
    ○ For non-ulcerative lesions eg nodules/papules/oedema/some ulcerative lesions
  3. Biopsy in case of surgery

GOLD standard: PCR (sensitivity 98%)
Other:
- Culture (sens 49%)
- Histopathology (sens 82%)
- Microscopy for AFB (sens 42%)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
588
Q

Current treatment for Buruli ulcer? Abx SEs?

A

Rifampicin 10mg/kg + Clarithromycin 15mg/kg DAILY for 8 WEEKs
In Australia: sometimes use Rif + Moxifloxacin *but no RCT

ALSO:
- Enhance wound healing: nutrition, wound care, skin grafting
- Disability prevention
- Rehabilitation

§ <2% patients
§ Skin rash (Rifampicin)
§ Anorexia/nausea/abdo pain/jaundice/renal failure (Rif)
§ Nausea/altered taste/jaundice/hepatitis (Clari)
§ Tendonitis, rash (Moxifloxacin)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
589
Q

A patient is treated for Buruli ulcer with 8 weeks of Rifampicin and Clari. After 2.5 weeks of treatment they return to the health centre with multiple nodules surrounding the original wound. What is your diagnosis? What causes this? How would you manage it?

A

Paradoxical reaction
- Multiple nodular lesions 2 weeks after start of Abx
- Inflammatory reaction that, prior to antibiotic treatment, is suppressed by mycolactone, the M. ulcerans toxin.
- As the organisms are killed by antibiotics, mycolactone production ceases and its suppressive effect is lost causing a rebound of inflammation
- Associated with high bacterial load

Can consider steroids but should heal on its own

Cover lesions with vaseline gauze + control lymphoedema with short stretch bandages +/- grafting

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
590
Q

Complications of buruli ulcer (eg untreated or late presented)

A
  • Contractures
  • Cancer
  • Amputation
  • Eye complications
  • Severe mental distress and reduced QOL
  • Burden on caregivers
  • Stigmatising
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
591
Q

Diagnosis for buruli ulcer is mainly via?

A

PCR

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
592
Q

What is podoconiosis? What causes it?

A

Steadily progressive foot and leg swelling (lymphoedema), often confused with lymphatic filariasis

Caused by triad of:
- Exposure to irritant soil
- Genetic susceptibility (link to SNPs in HLA Class II locus (particularly HLA DQA1) in Wellcome funded study)
- Foot vulnerability (lack of shoe-wearing and foot hygiene) + long term exposure

Rare in children, more common at higher altitudes and with Smectite in soil, farming communities + barefoot

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
593
Q

How do you diagnose podoconiosis? How does it present clinically?

A
  • Clinical
    ○ Hx, physical O/E, Ag and Ab tests to rule out lymphatic filariasis
  • Early signs
    ○ Itching, burning, splaying of forefeet, reversible plantar oedema, hyperkeratosis
  • Later signs
    ○ Persistent oedema, inter-digital maceration, nodules, ‘mossy’ changes, ‘waterbag’ or ‘fibrotic’ swelling
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
594
Q

Ddx of lymphoedema in tropical settings

A

Podoconiosis
Lymphatic filariasis
Onchocerciasis
Leprosy
Protein-energy malnutrition in children

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
595
Q

How can you tell the difference between lymphatic filariasis and podoconiosis?

A
  • Sea level: Podo>1500m, LF<1000m
  • Diagnosis: LF field ICT blood test, Podo clinical
  • First symptom site: LF not food, Podo toes/foot
  • LN attacks: LF precede swelling of limb, Podo follow swelling of limb
  • Chief site of swelling: LF above/below knee, Podo below knee
  • Prevention: LF mosquito nets/MDA, Podo: footwear/coverings/foot washing
  • Treatment: LF 2 drugs (albendazole/DEC) + foot hygiene, Podo: foot hygiene, bandages, socks etc
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
596
Q

Complications of Podoconiosis?

A
  • Bacterial co-infection (high rates of drug-resistance organisms found, mix gram neg + gram positive Bac)
  • Acute episodes (ADLA) characterized by malaise, fever, chills, diffuse inflammation and swelling of the limb, lymphangitis, adenitis and skin peeling
    ○ Require analgesia (e.g. paracetamol), fluids and antibiotic such as penicillin
  • Ulcer not responding to care -> refer to a surgeon for biopsy (?SCC/BCC)
  • Deep fungal infection. Additional swelling, on which small black dots may be visible -> refer to surgeon
  • Psychosocial and MH burden
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
597
Q

Principles of podoconiosis treatment

A

Hygiene
- BD, room-temp washing of limbs

Skin care
- Wash + dry + rub ointment/emollients, if breakages then washing + antibac/fungal creams, cover with sterile gauze for fly protection

Bandaging
- To reduce swelling, from toes to knee, V design, 5cm above upper limit of swelling (give people 2, one to wash and one to use)

Elevation and exercise
- Raise affected limb at night, ankle circles

Footwear
- Clean socks + feet, clean between toes, avoid tight shoes + low heel

Treatment of acute attacks
- Rest& elevate affected limb
- Coldwater immersion/cold soaked towels
- Anti-histamines + analgesia
- Encourage fluids
- Abx if no improvement in 24hrs
- Refer if pregnant, rigors, high fever, vomiting, confusion
- ?Associated diseases
DON’T: exercise, warm/heat on skin, open/cut blister or skin, bandage, rub herbs etc, use scarification

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
598
Q

What is Noma? What causes it, where is it found and what are some associated factors?

A

Rapidly acting orofacial gangrene
Affects kids aged 2-5 years old
14,000 cases yearly but now focused in South America, Africa and parts of Asia

Opportunistic infectioninte
Range of organisms found but not consistently (Fusobacterium necrophorum, Prevotella intermedia, Klebsiella, Streptococcus, Porphyromonas gingivalis, Campylobacter rectus, Treponema denticola)

Risk factors = No care access + socio-demographics + comorbidities
- Aged 2-5 years
- Chronic malnutrition
- Comorbidities
- Lack of access to quality healthcare including childhood vaccinations
- Not being breastfed
- Poor oral hygiene practices
- Low socioeconomic status of the family
High no. of previous pregnancies in mother

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
599
Q

Stages of Noma disease

A

0: Simple gingivitis
- Bleeding when touched or during brushing
- Red or purplish red gum
- Swelling gum

1: Acute necrotising gingivitis (indefinite duration)
- Spontaneous bleeding
- Painful ulceration of gum
- Ulceration with 1+ interdental papillae
- Fetid breath or halitosis
- Excessive salivation

2: Oedema (1-2 weeks)
- Rapid extension of gingival ulceration and mucosal tissue
- Fetid breath or halitosis
- Facial swelling or oedema
- Painful cheek
- High fever
- Excessive salivation
- Mouth soreness
- Difficulty eating
- Anorexia
- Lymphadenopathy

3: Gangrene (Progresses in 1-2 weeks)
- Excessive destruction of intraoral soft and hard tissue
- Lesion with well demarcated perimeter surrounding blackened necrotic centre
- Separation of slough -> leaves hole in face often around cheeks or lips
- Difficulty eating
- Perforation of cheek + exposition of teeth/denuded bones
- Progressive drying of facial gangrene, anorexia, apathy

4: Scarring
- Trismus (depending on lesion location)
- Sequestration of teeth and exposure of bones

5: Sequelae
- Disfiguring
- Trismus
- Teeth loss/displacement/dental anarchy
- Feeding + speech difficulties
- Fusion of maxilla and mandible bones
- Nasal regurgitation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
600
Q

Ddx for Noma?

A

> Tooth abscess
Cellulitis
Animal bites
Burn injuries
Syphilitic yaws
Buruli ulcer
Leprosy
Oral cancer
Chemical burns

Noma: will be quick destruction of facial tissue in a child with comorbidities, with well defined perimeter

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
601
Q

Treatment of Noma?

A

Abx
-> typical antibiotic regimen often includes:
Metronidazole (for anaerobes)
Penicillin or Amoxicillin (for aerobic bacteria)
Gent if gram neg

Wound debridement
Nutritional support
Reconstructive surgery
Physiotherapy
Psycho-social support

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
602
Q

WHO guidelines (condensed) for HBV treatment?

A

Cirrhosis -> treat with tenofovir or entecavir, if a child then with entecavir

No cirrhosis?
Basically persistently abnormal ALT + high VL -> treat
If VL <20,000 then defer and monitor
If ALT not that abnormal and/or lower VL -> defer and monitor

Should get regular screening for HCC/treatment response/toxicity (renal esp)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
603
Q

Why do HBV/HCV DNA/RNA levels become less helpful/not a good predictor of disease with hepatitis D co-infection?

A

HDV suppresses both HBV and HCV replication (DNA/RNA levels do not predict outcome)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
604
Q

Key facts hepatitis D?

A
  • Single stranded RNA
  • Can only infect with HBV (takes 3 proteins for viral envelope)
  • Transmission: blood/body fluids (MTCT is rare)
  • Epidemiology: 5% of HBsAg carriers (high seroprevalence in Middle East, Central Asia and South America and high-risk groups: IVDU, MSM, SW (a/w HCV, HIV) - up to 80%)
  • Incubation: up to 3/12
  • Course: usually found in screening of acute hepatitis or progressive chronic liver disease
  • Co-infection: acute self-limiting; chronicity rare
  • Superinfection: 5% acute liver failure; 80-100% chronicity; rapid progression (up to 80% cirrhosis in <10 years)
  • HDV suppresses both HBV and HCV replication (DNA/RNA levels do not predict outcome)
  • Diagnosis: serology (IgM/IgG) or molecular testing (RNA); guidelines for testing inconsistent (EASL: low HBV DNA and high ALT)

Treatment: IFN𝛼 <30% SVR at 6/12

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
605
Q

Treatment for chronic Hep C according to WHO?

A

DAAs usually 8-12 weeks, for all serotypes so genotypic testing not required eg
Sofosbuvir/Velpatasvir
Glecaprevir/Pibrentasvir

Good SVR (sustained virologic response) >95%: measured as virus is no longer detectable in the blood 12 weeks after completing therapy

If uncompensated cirrhosis may need longer course

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
606
Q

Stages of HIV infection and their respective relevant tests?

A
  1. Eclipse phase
    ○ Infection of mucosal tissue
  2. Viral stage I (viral RNA)
    ○ Viraemic
    ○ Test still negative
  3. Early Ag
    ○ P24 Ag
  4. HIV-specific Ab response
    ○ ELISA
    ○ Western blots
    ○ Partial control of viraemia but not complete
    ○ Chronic infection
    ○ Early chronic infection stage
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
607
Q

What is the window period for HIV testing? How does this relate to tests used for HIV?

A

Time between infection and test at which 99% of infections should be identified

4th/5th Gen: 45 days (4th gen median 18 days, IQR 13 - 24)

POCT/self-tests: 90 days

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
608
Q

If you are querying resistance in a HIV case where VL is undetectable what can you do?

A

Send pro viral DNA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
609
Q

What does reversion to wild type mean in the context of HIV infection?

A
  • Drug resistance mutations in HIV usually come at a cost to the virus, making it less “fit” in the absence of the drug. These resistant strains may replicate more slowly or less effectively compared to the wild-type virus.
  • When the selective pressure from the antiretroviral drugs is removed (for example, if the patient discontinues treatment), the wild-type virus can outcompete the drug-resistant strain because it is generally more fit.
  • As a result, the virus reverts to its original, drug-sensitive form.

Importance of Reversion to Wild Type:
* Clinical Implications: Reversion to wild type can be beneficial in terms of restoring sensitivity to the original drug, meaning that the same ART regimen may become effective again if resistance had previously developed.
* However, even though reversion occurs, archived resistance mutations can still persist in the viral reservoir and may re-emerge if the same drug is used again.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
610
Q

WHO clinical staging of HIV?

A

Primary HIV (CD4 ~ 350 - 500 cells/mm3)
- Asymptomatic, acute retroviral syndrome

Clinical stage 1 (CD4 ~ 350 - 500 cells/mm3)
Asymptomatic
Persistent generalised lymphadenopathy

Clinical stage 2 (CD4 ~ 350 - 500 cells/mm3)
Lots but Herpes zoster
Angular cheilitis
Recurrent oral ulceration
Papular pruritic eruptions (eosinophilic folliculitis)
Weight loss but less than 10% BW

Clinical stage 3 (CD4 ~ 200 – 350 cells/mm3)
Severe WL >10% BW
Persistent oral candidiasis
Oral hairy leukoplakia
Pulmonary tuberculosis
Severe bacterial infections (pneumonia etc)
Ulcerative stomatitis

Clinical stage 4 (CD4 <200 cells/mm3)
HIV wasting syndrome
Pneumocystis pneumonia
Oesophageal candidiasis
Extrapulmonary tuberculosis
Kaposi’s sarcoma
CNS toxoplasmosis
Extrapulmonary cryptococcosis
Disseminated non-TB mycobacterial infection
Chronic cryptosporidiosis
Chronic isosporiasis
Disseminated mycosis (dimorphic fungal)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
610
Q

Ddx for mediastinal lymphadenopathy?

A
  • TB
  • Lymphoma
  • Dimorphic fungal
  • Cryptococcosis
  • Nocardia
  • NTM
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
611
Q

When should you screen for cryptococcal disease? What to treat with if positive?

A

WHO now provisionally recommend screening all individuals with a CD4 count < 200 cells/µL

Treat patients without evidence of CNS disease with high dose fluconazole 1200mg for 2 weeks, followed by standard consolidation and maintenance therapy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
612
Q

Most common nerve (and palsy) affected by raised ICP? How does it present?

A

6th nerve palsy

Inward deviation of the eye (esotropia):
The affected eye turns inward toward the nose because the lateral rectus muscle is weakened or paralyzed, while the opposing medial rectus (which pulls the eye inward) is unopposed.

Diplopia (double vision):
Patients often experience horizontal double vision, particularly when looking toward the side of the affected eye. This occurs because the two eyes are misaligned.

Limited outward movement:
There is a marked reduction or inability to move the affected eye outward (abduction). For example, if the left 6th nerve is palsied, the left eye won’t move fully to the left.

Head turning:
To compensate for the double vision, patients may turn their head toward the side of the affected nerve (the side of the weakened lateral rectus muscle). This helps align the eyes and reduce double vision.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
613
Q

Presentation of cryptococcal meningitis

A

Difficult to differentiate between other causes. Headache/meningism/fever/focal neurology/abnormal mental status.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
614
Q

Common causes in adults and children of meningitis in HIV high prevalent settings

A

Adults: strep pneumoniae
Kids: Hemophilus influenzae, Strep pneumoniae, GNRs eg E coli/Klebsiella

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
615
Q

Textbook CSF findings TB infection:

A

Appearance: Clear/cloudy
Opening pressure: raised
WCC: raised (normally <500), lymphocytes
Protein: greatly raised
Glucose: low
Serum to glucose ratio: v low

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
616
Q

Textbook CSF findings viral infection:

A

Appearance: Clear
Opening pressure: normal/mildly raised
WCC: raised (normally <1000), lymphocytes
Protein: mildly raised, 0.5-1
Glucose: normal or slightly low
Serum to glucose ratio: normal or slightly low

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
617
Q

Textbook CSF findings bacterial infection:

A

Appearance: turbid/cloudy/purulent
Opening pressure: raised
WCC: raised, neutrophils
Protein: raised, >1
Glucose: low
Serum to glucose ratio: very low

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
618
Q

What is the commonest cause of dysphagia/odonophagia with oesophageal lesions in HIV?

A

CANDIDA
Treat but a lack of response should prompt endoscopy to investigate for:
* Cytomegalovirus
* Major aphthous ulceration
* Deep fungal infections
* Malignancy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
619
Q

Examples of coccidian parasites that can cause infection in HIV?

A

Cryptosporidium parvuum
* Zoonosis, contaminated water
* Most common cause of chronic diarrhoea
* No other treatment than ART

Cystisospora belli
- Widespread in environment
- Can cause refractory disease
- Treat with cotrimoxazole (or ciprofloxacin)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
619
Q

34 yo woman
Chronic diarrhoea, dehydration, profound weight loss
Newly diagnosed HIV, CD4 10
HIV wasting syndrome
* loss >10 % body weight
* diarrhoea >1 month
Ddx?

A
  • Cryptosporidium
  • Isosporiasis
  • Mycobacterium tuberculosis
  • Non-tuberculous mycobacteria – MAC
  • Cytomegalovirus
  • Microsporidiosis
  • Deep fungal infection eg cryptococcus, histoplasma
  • Lymphoma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
620
Q

How does chronic diarrhoea in HIV present for small vs large bowel and what are relevant pathogens?

A

Small:
- Bloating + abdominal pain
- Watery and large volume
* Cryptosporidium
* Isospora
* NTM
* HIV enteropathy
* Microsporidiosis
- Ix: Endoscopy and biopsy

Large:
- Small volume, blood/mucus/WBCs
- Tenesmus
- CMV, TB, Amoebiasis, Schisto

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
621
Q

When are patients at highest risk of severe bacterial infections with HIV? What are some common bugs?

A

First 3 months of ART and if not on ART. Salmonella (typhoidal and non-typhoidal), Pneumococcus, E Coli.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
622
Q

When would you treat with steroids for TB related conditions?

A

TBM -> if HIV negative / if SOL
TBM -> consider if HIV positive
Inflammatory pericarditis
Severe and neurological TB IRIS

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
623
Q

How do you differentiate between Toxoplasmosis and brain abscess on CT?

A

You can’t. Would need micro. Toxo treat with co-trimox.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
624
Q

What virus could be found in CSF in a HIV patient with CNS lymphoma?

A

EBV

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
625
Q

A HIV patient has a solitary lesion on neuroimaging and has not responded well to co-trimoxazole. What should be considered as another differential? How would you confirm this?

A

Consider CNS lymphoma
Brain biopsy for definitive diagnosis
Treat with chemo/radio/ART

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
626
Q

Key facts about progressive multifocal leukoencephalopathy

A

· Caused by JC virus, insidious onset, occurs at CD4 counts <100 cells/µL
· Progressive focal neurology (mainly motor)
· Normal CSF (JC virus PCR usually positive), MRI is diagnostic

No known effective treatment, give ART

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
627
Q

Histoplasmosis
- Presentation?
- Ix
- Treatment

A
  • Progressive disseminated infection
  • Fever, fatigue, weight loss, and hepatosplenomegaly
  • Cough, chest pain, and dyspnea in ~ 50% of patients
  • CNS, GI, and cutaneous manifestations in a smaller
  • percentage of patients
    If CD4 counts >300 cells/mm3, histoplasmosis is often limited to the respiratory tract with cough, pleuritic chest pain, and fever

Ix:
- Microscopy - characteristic 2 to 4 µm in diameter budding yeast
- Culture (lysis-centrifugation technique) blood, bone marrow, respiratory secretions
- PCR
Antigen detection (blood or urine) by EIA of LFA

Mild: itraconazole

Severe/CNS: induce with ambisome then itraconazole maintenance (can be lifelong eg for AIDS pts)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
628
Q

CMV in HIV patients
- Presentation
- Diagnosis
- Treatment

A

Disseminated or localized end-organ disease in individuals who have very advanced HIV disease (CD4 <50 cells/µL) – Retinitis, colitis, pneumonitis, encephalitis

Diagnose with PCR

Treat with valganciclovir

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
629
Q

Most common malignancies in cancer?

A
  • Cervical cancer
  • Kaposi’s sarcoma
    ○ Mortality at 8 months after KS -> poor outcomes
  • Lymphomas (NHL)
  • Castleman’s disease (Lymphoproliferative disorder, unicentric vs multicentric)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
630
Q

Impact of CD4 deletion on TB pathophysiology in HIV?

A

○ Less CD4+
○ Alteration in macrophage activation (less IFN-Y production and in turn, granuloma formation)
○ Alteration to neutrophils, less epithelioid cells, less Langhans cells
○ Less effective granuloma formation
○ Altered adaptive cell populations + altered phenotypes of cells

Advanced: increased necrosis + increased neutrophils + increase Mtb => granulomas poorly organised or absent

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
631
Q

Strategies for HIV and TB interventions for control

A

2000: DOTs
Mid 2000s: HIV control
2005ish: HIV/TB Collaboration activities
2010ish: 3 ‘I’s (intensive case finding/IPT/infection control)
2012: HIV/TB policy updates eg early ART, WHO endorsed Xpert
2014ish: End TB strategy, UNAIDS 90-90-90, WHO test can treat
2020: UNAIDs 95-95-95

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
632
Q

When might you trial antibiotics as a mode of TB diagnosis?

A

LMICs
- Bacterial LRTI is important differential for pulmonary TB
- Assumption that LRTI improves with short course of Abx
○ PTB will not improve with antibiotics
○ Widely used intervention (especially low resource settings)
○ Recommended in several TB diagnosis guidelines
○ Little consensus on which Abx/duration
Sensitivity ~70%, Specificity ~77%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
633
Q

Should you give 4 month treatment regimen with Rifapentine to TB patients living with HIV?

A

Rifapentine based 4 month regimens are in WHO guidelines for CD4 counts >100
(only 8% of trial participants PLHIV)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
634
Q

What is the difference between rifampicin and rifapentine?

A

Rifampicin: Shorter half-life, used daily, preferred for active TB treatment.

Rifapentine: Longer half-life, used weekly or biweekly in latent TB treatment or intermittent regimens, with fewer dosing requirements.

Both can cause orange discolouration of body liquids. Both inhibit RNA polymerase.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
635
Q

Which rifamycin class Abx has less interactions with ARTs so might be used more safely for TB in people living with HIV?

A

Rifabutin
- Less potent CYP450 inhibitor
- Can cause Uveitis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
636
Q

Who can you consider TPT = TB preventative therapy in? What are some issues with it? What are the preferred therapies?

A
  1. People living with HIV (PLHIV), due to their significantly higher risk of developing TB.
  2. Close contacts of active TB cases, especially children under 5 and immunocompromised individuals.
  3. Immunocompromised individuals, such as those on immunosuppressive drugs, or organ transplant recipients.
  4. People with silicosis, as this condition increases TB risk.
  5. Children under 5 years old, who are at high risk after exposure to TB.
  6. Healthcare workers and other high-risk populations, such as prisoners or people in homeless shelters.

Issues: resistance/poor adherence/pill burden/availabiltiy

Examples:
1st: Isoniazid/Rifapentine weekly 3 months OR Isoniazid/Rifampicin daily for 3 months
2nd: Monotherapy eg Isoniazid daily for 6-9 months

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
637
Q

Generalised life cycle of coccidia?

A

Oocyst ingested
Asexual reproduction leads to schizogony repeated
OR
Sexual reproduction leads to gamete formation
Release of oocyte
Resistent oocytes (ie with outer coat) are shed in faeces
Oocyte maturation (sporulation, sporogony) normally outside the body

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
638
Q

What do mature (sporulated) coccidian oocysts contain?

A

Sporocyst
Sporozoites
Residual body

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
639
Q

Life cycle of cryptosporidium?

A
  • Oocyst Stage (Infective Stage): The Cryptosporidium parasite starts as a resistant oocyst (a spore-like form) in contaminated food, water, or surfaces. These oocysts are ingested by a host, such as a human or animal.
  • Excystation in the Intestine: Once inside the host’s stomach or small intestine, the oocyst releases infectious forms called sporozoites. This process is called excystation.
  • Invasion of Intestinal Cells: The sporozoites attach to and invade the epithelial cells of the intestinal lining. Inside these cells, they undergo asexual reproduction, forming meronts.
  • Asexual and Sexual Reproduction: Type I meronts release merozoites which invade new cells for further asexual reproduction, type II meronts release merozoites which undergo sexual reproduction to form microgamonts (male gametes) and macrogamonts (female gametes). These gametes fuse to form a zygote.

Formation of New Oocysts: The zygote develops into a new oocyst, if its thin-walled it can stay inside the host -> autoinfection. If thick walled it exists host, shed in faeces.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
640
Q

Clinical presentation of cryptosporidium?

A
  • Wide ranging s/s: asymptomatic (common) -> life threatening
  • Watery diarrhoea is most frequent symptom
    ○ + dehydration, WL, abdo pain, fever, N&V
  • Immunocompetent lasts 1-2 weeks
    ○ Immunity to reinfection is incomplete -> risk of multiple episodes
  • Chronic and more severe in immunocompromised -> infection spreads from intestine to hepatobiliary and pancreatic ducts -> cholangiohepatitis, cholecystitis, choledochitis or pancreatitis
  • Risk of respiratory cryptosporidiosis?
    ○ Known in immunocompromised
    ○ New data suggesting in immunocompetent children in Malawi via cryptosporidial diarrhoea and unexplained cough, transfer via aerosolised droplets or contact with fomites contaminated by coughing

Long term morbidity (up to 10 years)
Symptoms can persist following infection eg weight loss, blood in stool, chronic diarrhoea - IBS like symptoms

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
641
Q

Is cryptosporidiosis in infants in less-developed countries a significant cause of
malnutrition, growth impairment and cognitive deficit? Is cryptosporidiosis associated with anything else?

A

Cryptosporidiosis interacts in a complex way with malnutrition and the intestinal microbiome to cause long-term harm to young children in less-developed countries

  • A change in intestinal cell kinetics, leading to fewer mature enterocytes and impaired nutrient uptake, may
    be one significant reason (Wallbank et al. 2024)

Other associations with bowel cancer -> higher rates of infection in BC compared to controls

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
642
Q

How do you diagnose Cryptosporidiosis?

A
  • Light microscopy of faecal specimens - staining with Ziehl–Neelsen, Auramine or Safranin (last not quite so good)
    • MUST measure (5-5.5 µm) and check bright red (Z-N)
  • Direct FAT and immunochromatographic RDTs available and widely used
    • But note: the mean sensitivities of the three best kits tested, by species, was C. parvum 73.5%; C. hominis 80.8%, other species 27.7% (Agnamey et al. 2011)
    • The sensitivity and specificity of 3 antigen detection kits, Auramine and Immunofluoresence were good and all were better than modified Ziehl–Neelsen (Chalmers et al. 2011a)
  • PCR for diagnosis and genotyping
    PCR based methods appear to have the highest sensitivity and specificity - for a comparison of four commercial and four in-house protocols (including performance with some of the less-common species) see Costa et al (2021)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
643
Q

Size of cryptosporidium?

A

5micrometers, cherry red

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
644
Q

How to differentiate cryptosporidium and cyclospora?

A

Cryptosporidium: 5micrometers, cherry red
Cyclospora: 5-8micrometre ie bigger, can be red, acid-fast behaviour

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
645
Q

Criteria for identifying cyclospora?

A

Criteria for identifying oocysts:
– Usually round
– Small, normally about 8μm diameter
– Dark, refractile wall, thicker than most other gut protozoan
cysts
– “Broken glass” contents

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
646
Q

In which new group is PCP incidence rising? How does pathophysiology compare to HIV patients?

A

solid transplant recipients and autoimmune inflammatory diseases

These patients tend to have a sudden acute onset and worse survival outcomes than HIV+
patients -> ie HIV patients have higher fungal loads but poor immune system so less mortality, whereas organ transplant patients etc have lower fungal loads but better immune system so mount more fatal responses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
647
Q

What is the pathophysiology of PCP?

A

Organism adheres to Type 1 pneumocytes lining alveoli
Seems to be immune component involved

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
648
Q

Black on green lung on microscopy slide - what is organism and stain?

A

Pneumocystic cysts -> Grocott Silver stain

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
649
Q

How do you treat PCP?

A
  1. Co-trimoxazole
    ○ High incidence of SEs in HIV+ patients
    ○ Mutations in sulphonamide target enzyme might reduce effectiveness (conflicting evidence)
  2. Other
    ○ Pentamidine
    ○ Dapsone-trimethoprim
    ○ Clindamycin-primaquine
    ○ Atovaquone
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
650
Q

How do you diagnose PCP?

A
  • Need to distinguish between colonisation and actual PCP
  • BAL (best sample)
    ○ Invasive but more sensitive than induced sputum
    ○ Staining (silver; direct FAT more sensitivie)
    ○ PCR better than microscopy
    ○ Metagenomic sequencing
  • Quantitative PCR using oral wash samples
  • Some DNA has been detectable in plasma of AIDs/PCP patients but no method really used
  • Detection of (1,3)-beta-D-Glucan: less invasive
    ○ Sensitivity high but not as good as PCR, especially when used alone
    ○ Specificity: same polysaccharide present in cell walls of other fungi/serum of patients who have had certain procedures etc
    Performs worse if lower fungal load
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
651
Q

What is the Epi of PCP?

A
  • Host-specific ie human parasite
  • Airborne transmission
  • Transplacental transmission - rabbits, maybe humans
  • Very widespread in air -> ie most kids colonised before school
  • Probable later attacks of PCP in immunocompromised are NEW infections not reactivation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
652
Q

How do you diagnose microsporidiosis?

A
  • Staining and light microscopy (speciation difficult or impossible)
  • Electron microscopy (define species)
  • Nucleic acid amplification methods (increasingly important)
  • In situ hybridisation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
653
Q

What pathogens cause leprosy? What are some facts about them that make the diagnosis difficult?

A

Mycobacterium leprae and Mycobacterium lepromatosis

  • Intracellular organism
  • Not possible to culture
  • Long incubation period
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
654
Q

Transmission of leprosy, who does it effect?

A
  • Unclear
  • ?similar to TB, ie aerosol spraying into nasopharyngeal tract, coughing etc
    ○ Bacilli found in nasal droplets of highly infected people and thought to enter body through upper resp tract
  • Affinity for cooler sites of body -> peripheral nerves and skin
  • More rarely: eyes, mucous membranes, testes, bones and viscera
  • Disease:
    ○ Long incubation period, 3-5 years, may vary from 6 months to 20 years
    ○ Progress of disease depends on immunological status of person
  • Animal reservoirs:
    ○ Armadillo?
    ○ Some monkeys/squirrels
  • Has been found in soil and can survive in it - ? Environmental pathogens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
655
Q

Presenting complaints of leprosy? What type of skin lesions exist?

A
  • Numbness /loss of function in hands and feet
  • Deformities in hands and feet
  • Painless ulcers or burns (but might smell)
  • Nasal stuffiness
  • Weakness in eyelids
  • Reaction presentation: fever, joint pains, inflammed skin
  • Skin lesions - 95% patients

Skin lesions:
○ Tuberculous end lesions are asymmetrical and well defined but there is some anaesthesia ie loss of sensation

○ Lepromatous lesions more symmetrical, widespread, at worst -> nodules and infiltration

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
656
Q

Skin signs and nerve signs in leprosy?

A

Skin lesions:
○ Tuberculous end lesions are asymmetrical and well defined but there is some anaesthesia ie loss of sensation
○ Lepromatous lesions more symmetrical, widespread, at worst -> nodules and infiltration

  • Enlarged nerves
  • Dryness in hands and feet
  • Loss of sensation in hands and feet
  • Painless ulcers and wounds
  • Weakness in motor function in hands and feet (progression to motor nerves being affected)
  • Deformities in hands and feet
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
657
Q

Investigations to diagnose leprosy?

A
  • Slit skin smear for AFB
    ○ Suspected lesions and sites commonly affected eg forehead, eyebrows, ear lobes) are sampled
  • No useful serological or skin test
  • Skin and nerve biopsy -> granumolas?

Bacteriological index
- ZN staining of smears made from skin slits
- Read on logarithmic scale 0-6
- Useful in lepromatous cases and unusual skin lesions
- Can start treatment where clear-cut skin and/or nerve lesions are present before skin slit smears results available
- Negative skin-slit smears do not exclude diagnosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
658
Q

WHO cardinal signs of leprosy?

A

Definite loss of sensation in a skin lesions consistent with leprosy
Thickening or one or more peripheral nerves
Skin smear or biopsy positive for AFB

If these then start Tx

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
659
Q

WHO classification of leprosy?

A
  • Paucibacillary = 5 lesions or less
  • Multibacillary = 6 lesions or more

If skin smear positive -> patient automatically classified as MB regardless of no, of patches

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
660
Q

What is the treatment of leprosy? How is it changing? What are the side effects?

A

PB
- 6 months treatment
- 2 drugs:
○ Rifampicin once a month
○ Dapsone daily
MB
- 12 months treatment
- 3 drugs:
○ Rifampicin once a month
○ Dapsone daily
○ Clofazimine daily

NOW WHO recommends to give ALL patients the 3 drug packet but 6 months vs 12 months according to two groups. Enables just a single packet to be given. Patients don’t like it as Clofazimine causes hyperpigmentation so if they have PB then why are they taking drug?

SIDE EFFECTS
Dapsone:
- Anaemia
- Hypersensitivity syndrome (fatal in 10%)
Clofazimine
- Hyperpigmentation
- Ichthyosis (dry scaly skin)
Rifampicin
- Orange urine/tears: 1-2 days only

Safe in: pregnancy, breast feeding, HIV

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
661
Q

Management of household contacts of leprosy?

A
  • Increased risk of developing leprosy -> need prolonged contact with person and they probably need high BI
  • WHO recommend chemoprophylaxis with single dose of rifampicin to contacts
    ○ Household
    ○ Household and neighbours
    ○ Communities
  • Screened for evidence of clinical leprosy and TB

Now new drugs being considered for contacts eg adding clarithromycin and treating for a couple of months.

  • Brazilian ministry of health recommends all household contacts be given two doses of BCG
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
662
Q

Types of adverse reactions + their features

A

Type 1 reactions
Features:
Can occur at any point in journey
Skin and nerve acute inflammation
Typically effects pre-existing skin lesions
Presents as large skin lesions/Scaling/Oedema/Annular lesions
Associated with facial and peripheral oedema
Nerve inflammation may lead to permanent loss of nerve function

ENL
Multisystem disorder -> can occur anytime and can be recurrent or chronic
Can present with any systemic inflammatory symptoms eg fever/malaise/oedema/any -itis type
Effective treatment: Thalidomide (but issues with access/restriction/banning etc) so patients often given other drugs eg prednisolone, Clofazimine, Azathioprine

Silent neuropathy

Lucio’s phenomenon -> Widespread ulcerative lesions in patients with high bacillary load who likely are not on treatment or who have been under-treated. Start MDT immediately
Consider steroids or other anti-inflammatories
Wound care/Abx if secondary infections

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
663
Q

Corticosteroid adverse effects:

A
  • Lipodystrophy
    • Infections particularly strongyloidiasis
    • Hypertension
    • DM
    • Peptic ulceration
    • Mood disturbance
    • Osteoporosis
      • Cataracts
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
664
Q

How are nerves effected in leprosy?

A
  • Type 1 reactions -> Nerve inflammation may lead to permanent loss of nerve function
  • Nerve enlargement -> Increased risk of entrapment + compression
  • Vascular effect
  • Direct effect of mycobacterium on nerve tissue
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
665
Q

A patients with painful necrotising skin lesions to leprosy clinic. Skin biopsy shows vasculitis with thrombosis (blood clotting) and extensive bacillary infiltration. What has happened? How would you treat?

A

Lucio’s phenomenon

Start MDT immediately
Consider steroids or other anti-inflammatories
Wound care/Abx if secondary infections

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
666
Q

Primary and secondary impairments of leprosy

A

Hand/feet:
Primary
Loss of sensation, sweat
Muscle weakness

Secondary
Claw fingers/toes/wrist/foot drop ->
Contracture
Ulceration
Loss of digits

Eyes:
Primary
Lagophthalmos
Corneal anaesthesia

Secondary
Exposure keratitis
Corneal ulcer
Blindness

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
667
Q

Interventions to reduce disability in leprosy?

A
  1. Early case detection
    Helps also pick up paediatric cases and G2D. Allow for reliable burden estimation, district prioritisation and identifying populations at risk.
  2. Self-care practice
    Skin care, wound care, exercise, footwear, health education + peer support groups
  3. Footwear
  4. Psychosocial support (PSS)
  5. Reconstructive Surgery
    need nerve assessments to find patients at risk ie muscle strength, sensation and sweating
    correct wrist/foot drop, decompress nerves, collapse nose, eye surgery
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
668
Q

Spectrum of disease in leprosy and relevant pathophysiology?

A

Only 5% infected with leprosy bacilli will develop the disease
Indeterminate: detectable but no clinical manifestations

Tuberculoid: strong immunity, few bacteria

Lepromatous: weak immunity, many bacteria
- Generalised -> nerves, skin + other tissue
- Macrophages ingest but don’t digest the bacilli due to lack of cell mediated immunity (CMI) -> convenient host
- Multiplication of bacilli and transport by macrophages to other parts of body

Upgrading towards tuberculoid end = not necessarily a good thing as over activity of T cells -> inflammation -> tissue damage

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
669
Q

Clinical spectrum of skin lesions in leprosy and features? Name of classification?

A

TT Tuberculoid leprosy: well defined (asymmetrical, definite anaesthesia)

BT Borderline Tuberculoid: ill-defined satellites (asymmetrical, definite anaesthesia)

BB Borderline borderline: circinate (asymmetrical, definite anaesthesia)

BL Borderline lepromatous: hypoesthetic patches (symmetrical + variable lesion anaesthesia)

LL Lepromatous: Nodules and infiltration (symmetrical + variable lesion anaesthesia)

RIDLEY JOPLING CLASSIFICATION is the 5 things
Now use WHO criteria instead

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
670
Q

Eye issues in leprosy?

A

○ Lagophthalmos
○ Decreased corneal sensation
○ Acute/chronic iritis
○ Cataract

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
671
Q

Nerves to examine in leprosy?

A

Supraorbital
Great auricular
Cervical
Radial
Median
Ulnar
Radial cutaneous
Common peroneal
Posterior tibial

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
672
Q

WHO disability grading for leprosy components?

A

Eyes/Hands/Feet
Grade 0,1,2

Grade 2 disability suggests visible functional disabilities in the person and he will not only be economically affected by these but also more at risk of stigmatisation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
673
Q

Basic framework for diagnosing leprosy?

A

History
Full examination of skin, nerves, deformities, ulcers, wounds
ST (sensory testing)
VMT (voluntary muscle testing)
Disability grading including eyes
Skin slit smears/biopsy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
674
Q

Two medically relevant families of snakes and basic features?

A
  1. Viperidae
    ○ Vipers (eg western Russell’s viper, pit viper), adders, pit-vipers, moccasins, rattlesnakes
    ○ Relatively short thick body, short tail, distinctive dorsal pattern, slow-moving ambush predators but strike like lightening
  2. Elapidae
    ○ Cobras, king cobras, kraits, mambas, coral snakes, all Oceanian/Australian venomous snakes and sea-snakes
    ○ Relatively long thin body, long tail, uniformly coloured (except corals), fast, some spread good
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
675
Q

Four main families of snake venom toxins?

A

○ Phospholipases A2
○ Metalloproteases
○ Serine proteases
○ Three-finger toxins (neuro, cyto toxins)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
676
Q

Factors influencing EPI of snakebites

A
  • Depends on interaction with snakes and their readiness to strike
  • Meteorology: heats, rains, flooding
  • Cycles of diurnal and seasonal activity:
    ○ Snake: nocturnal hunting + mating season
    ○ Human: occupational (agriculture, pastoralism), building projects, travel
  • Sleeping on the ground
    ○ Nocturnal krait bites in South Asia
  • Intentional snake handling
    ○ Restaurants/leather
    ○ Performance
    ○ Religion/Science
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
677
Q

What are dry bites for snakes?

A
  • Cause puncture marks on skin indicating penetration by teeth or fangs
  • BUT results in no evidence of local or systemic envenoming over next 24hr (no venom injected)
  • Incidence varies with snake species:
    ○ Saw-scaled vipers (Echis): <10% bites are dry
    ○ Australian brown snakes (Pseudonaja): >80% of bites are dry

Mechanism unknown (?mechanical disadvantageous defensive strike, ?snake-controlled metering)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
678
Q

What are two examples of drugs based on reptile venom toxins? What reptiles?

A

ACE-I: jacaraca snake in Brazil

GLP-1: Toxin from North American Gila monster

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
679
Q

What are the effects of envenoming per system and associated snake families?

A

LOCAL
Cytotoxicity (some cobras and most vipers)
- Local swelling
- Bruising
- Necrosis
- Risk of bacterial infection

Systemic
Haemotoxicity (vipers and oceanic/australian elapids)
- Coagulopathy
- Bleeding
- Extravasation

Neurotoxicity (elapids, a few vipers)
- Descending flaccid paralysis

CVS toxicity (vipers)
- Arrhythmias
- Myocardial damage
- Capillary leak
- Hypovolaemia
- Shock

Myo-toxicity
- Generalised rhabdomyolysis
- Hyperkalaemia

Nephro-toxicity
- AKI
- Pigment nephropathy
- Microangiopathic haemolysis
- Thrombotic microangiopathy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
680
Q

How can CVS toxicity result from snake bites?

A
  • Hypovolaemia from extravasation into bitten limb
  • Generalised increase in capillary permeability
  • Direct myocardial toxicity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
681
Q

Causes of symptoms in snakebites?

A

Anxiety -> hyperventilation, conversion disorders

Pre-hospital treatment

Pre-existing medical conditions and regular medical conditions

Secondary bacterial wound infections eg tetanus, necrotising faciitis, septicaemia

Secondary hypoxic damage resulting from delayed resuscitation of venom - induced shock or respiratory failure

Then finally envenoming

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
682
Q

Test for consumption coagulopathy as a result of snake venom?

A

WHOLE BLOOD CLOTTING TEST
- Bedside test for 20mins for coagulopathy
- Few ml of venous blood in new, clean, dry (soda) glass vessel
- Leave undisturbed for 20mins at ambient temp
- Tip once
- Positive -> non-clotting, incoagulable blood if blood remains liquid like water
- Negative -> clotting, coagulable blood if blood clots
- Repeat with normal control if result inconsistent with clinical status

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
683
Q

What does pressure pad immobilisation aim to do in snakebites?

A

Aim to compress veins and lymphatics in immediate vicinity of bite -> draining depot of injecting venom

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
684
Q

First aid and treatment of snakebite? Indications for antivenom?

A

FIRST AID
1. Remove from danger and reassure
2. Immobilise whole body, especially bitten limb (need to prevent muscle contraction)
3. Remove anything tight eg bracelets, anklets etc from limb
4. Apply pressure-pad-immobilisation immediately
5. Prevent shock, airway obstruction and resp failure in transmit, oxygen (?atropine + neostigmine for cobra bite paralysis)
6. Discourage traditional treatment
7. Get to hospital

Treatment
1. ABCDE + oxygen intubation

  1. Admit all proven or suspected snakebite cases for min of 24hr observation to cover late evolution of symptoms of envenoming
  2. Assess for signs of local and systemic envenoming (including bedside test for coagulopathy) ?ANTIVENOM (SAME DOSE for kids and adults)
  3. Try to diagnose species of snake
  4. Analgesia - paracetamol/acetaminophen
  5. Tetanus toxoid booster to all cases (all bites are penetrating injuries and injection provides reassurance)
  6. Watch for secondary bacterial wound infections (potentially fatal tetanus, nec fasc, septicaemia)
  7. Consider IVF/intubation + ventilation/RRT

ANTIVENOM indications:
- Shock
- Spontaneous systemic bleeding +/- incoagulable blood
- Neurotoxicity (ptosis or more)
- Black urine (haemoglobin or myoglobin)
- Rapidly progressive local swelling (especially bites on digits)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
685
Q

What are the different types of antivenom?

A

Monovalent: cover one species

Polyvalent: cover multiple medically important species in the region eg Indian Polyvalent ASVs cover for big 4

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
686
Q

How do you dose antivenom in children as compared to adults?

A

SAME INITIAL DOSE

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
687
Q

Surgical mx of snake bites?

A
  • Drain abscesses
  • Aspirate large tense blisters using fine needles
  • Debride/amputate necrotic tissue early (beware of nec fasc) and apply skin grafts
  • AVOID fasciotomy (s/s of raised compartmental pressure not accurate in snake bite
  • Encourage early rehab
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
688
Q

Treatment of scorpion stings? What a trick to prevent being bitten by them?

A

Treatment of local pain:
Infiltration/digital block with 1% lignocaine or something similar

Treatment of systemic envenoming:
Rare severe cases: may require ICU/CV/resp monitoring for minimum 24hr (ECG/echo)

Antivenom for:
- Any systemic symptoms/signs not explained by pain/anxiety alone, especially in small children
- Administration: slow IV injection/infusion

Use UV light to show them

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
689
Q

Symptoms of fish stings?

A

Local:
- Intense pain, tissue necrosis, secondary marine/aquatic bacterial infections (Staph, Strep, Vibrio vulnificus, Mycobacterium marinum, Aeromonas hydrophila)

Systemic:
- N&V, fever, hypotension, bradycardia, other cardiac arrhythmias, reps distress, pulmonary oedema, delirium, convulsions, autonomic stimulation, paralysis
- Rare cardiac arrest - reef stonefish Synanceia verrucosa is most dangerous

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
690
Q

Key examples of stinging fish?

A
  • Stone fish (Synanceia)
    • Scorpionfish (Scorpaenidae)
    • Lionfish
    • Catfish
    • Toadfish
    • River ray
    • Sting ray (Potamotrygon)
    • Dogfish
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
691
Q

Apart from fish stings, what are two families that have genus/species that can cause marine stings?

A

Echinoderms eg star fish or sea urchins

Cnidarians (Coelenterates) eg jellyfish etc

692
Q

What are some features of sting by Echinoderms?

A

STAR FISH OR SEA URCHINS
○ Venomous spines and grapples (pedicellaria)
○ Severe local pain, redness, swelling, blue/violet staining
○ Vomiting, rare systemic effects (weakness, paraesthesia, cardiac arrhythmias)
○ Marine bacterial wound infection

693
Q

What are some examples and features of sting by Cnidarians?

A

○ Surface of cnidarians (esp tenticles) are studded with millions of cnidocytes

○ Contain explosive nematocysts extruding stinging tubes for predation

○ Deadly jellyfish for example:
§ Australian box jellyfish (Chironex fleckeri) “sea wasp” - up to 3m long
□ Features:
® Intense local pain
® Cough, N&V, abdominal colic, diarrhoea, rigors
® Severe MSK pains
® Syncope, profuse sweating, cyanosis, generalised convulsions, pul oedema, rapid death

§ Nomura’s jellyfish (Nemopilema (Stomolophus) nomurai

§ Irukandji (Malo kingi) - around 1cm long, has lots of stinging tubes
□ Features:
® Minimal local signs
® Delayed (5-50mins) severe low back, “boring”
® Muscle pain/cramps limbs, trunk - in waves
® Chest tightness
® Sweating, restlessness, anxiety, resp distress, pul oedema, potentially fatal acute Takotsubo-like cardiomyopathy
® HTN causing fatal intracerebral haemorrhage
® ?hypercatecholaminaemia

§ Look out for local pain, wheals (indiagnostic patterns), swelling, erythema, vesiculation, necrosis

694
Q

What type of disease is Leishmaniasis?

A

Kinetoplastid parasitic disease -> flagellate protozoa

695
Q

Anthroponotic cause of visceral leishmaniasis?

A

L. donovani

696
Q

Causes of visceral leishmaniasis?

A

Visceral leishmaniasis a.k.a kala azar
Leishmania donovani
Leishmania infantum

Post Kala azar Dermal
Leishmaniasis (PKDL)
Leishmania donovani

697
Q

Causes of cutaneous leishmaniasis?

A

Cutaneous leishmaniasis
* ”localised”
L. major, L. tropica, L.mexicana, L. braziliensis

  • Mucosal lesions
    L. aethiopica
  • Diffuse (DCL)
    L. mexicana, L aethiopica
  • Recidivans
    L. tropica
  • Mucocutaneous leishmaniasis
    L. Viannia sub-genus
698
Q

Case definition of visceral leishmaniasis?

A

about 2-4 months after infection
at least 2 weeks of fever (rule out Malaria)
enlarged spleen and liver Ⓒ spleno- or Ⓓ hepato- megaly)
weight loss (cachexia)
Anaemia
+ve test (non-invasive)

699
Q

Life cycle of leishmaniasis?

A
  1. Infection of Sandfly (Promastigote Stage):
    • Female sandfly takes blood meal -> ingests macrophages that contain the amastigote form of the Leishmania parasite
    • Inside the sandfly midgut, the amastigotes transform into promastigotes, which are the motile, flagellated form of the parasite
    • Promastigotes divide + multiply in the sandfly’s midgut + eventually migrate to the fly’s proboscis (the biting mouthpart)
  2. Transmission to Humans:
    • When the infected sandfly bites a human, it injects the promastigotes into the skin.
    • The promastigotes are then phagocytosed (engulfed) by macrophages in the human’s skin.
  3. Amastigote Stage in Humans:
    • Inside the macrophages, the promastigotes transform into amastigotes, the non-motile, intracellular form of the parasite.
    • The amastigotes multiply within the macrophages, causing the cells to rupture and release new amastigotes into the bloodstream or nearby tissues, where they infect more macrophages.
  4. Infection of New Sandflies:
    When a sandfly bites an infected human or animal, it ingests macrophages filled with amastigotes, and the cycle begins again.
700
Q

POST-KALA-azar DERMAL LEISHMANIASIS facts

A
  • Seen in East Africa and South Asia when a VL infection has been clinically cured (eg L.donovani)
  • Can also occur after L.infantum VL infection
  • Parasites are localised in the skin, within lesions which range in appearance from slight to severe
  • Some differences in PKDL presentation between East Africa and South Asia
  • RESERVOIR for infection -> ie sandflies can bite the skin
  • South Asia
    ○ Macular
    ○ After VL: 10-30%
    ○ 0-3 years after VL
  • E Africa
    ○ Papular
    ○ After VL: 50-60%
    ○ 0-13 months after VL
701
Q

Features of cutaneous and mucosal leishmaniasis?

A

CUTANEOUS
- Localised or simple CL
- Eg L.major, L.tropica, L.mexicana, amazonensis
- Single or limited number of discrete lesions
- Self culture in months-years
- DTH+
- Disfiguring

MUCOSAL
- Mostly L.aethiopica infections
- Also seen with L.infantum, L.major and L.tropica
- Surface lesions of oral mucosa
- Not structurally destructive

702
Q

Features of diffuse cutaneous leishmaniasis?

A

L. aethiopica and some L. m. amazonensis infections

Multiple, coalesced lesions. Non-ulcerative, +++amastigotes, (“anergy”)
+++Leishmania antibodies

General T cell hypo-responsiveness (DTH –ve)
* No macrophage activation to clear parasites or induction of cell-mediated immunity
* Absence of IFN-γ allows multiplication and progression of lesions

Poor prognosis as highly refractory to treatment, immunotherapy +conventional treatment more successful

703
Q

Features of LEISHMANIASIS RECIDIVANS?

A
  • Rare
  • Relapsing L. tropica, L major or L. braziliensis
  • Develops after an LCL infection has been resolved, months to years later. Close to the original lesion or at site of trauma (LATENCY)
  • Amastigotes very sparse - PCR diagnosis
  • Difficult to treat
704
Q

Interactions of visceral leishmaniasis and other infections or conditions?

A

HIV
- two exacerbate eachother -> VL if HIV+ promotes progression of HIV/AIDS
- Asymptomatic or latent VL will recrudesce on HIV infection
↓CD4 cells leads to ↓IFN-Ɣ leading to ↓ cell-mediated response

TB
More patients presenting with TB + VL
Exacerbate each other
(Some TB+VL+HIV)

HELMINTHS
Worm infections induce a regulatory immune response → control of Leishmania
(Murine model of S. mansoni and L.
donovani and L. major)

PREGNANCY
* Associated reduced cellular immunity may result in VL exacerbation.
* Reports of transplacental transmission
* Pregnancy has a higher risk of early VL relapse.
* Newborns infected with Leishmania can have delayed symptoms

705
Q

Cellular biology of leishmaniasis?

A
  • Digenetic single-celled organism ie exists in 2 forms
  • ~1-2 um diameter
  • No flagellum
  • Obligate intracellular amastigote in humans
    ○ Parasitophorus vacuole pH 5.5
    ○ Amastigotes can survive in their vacuole -> have own protein pumps etc
    ○ Macrophages don’t “see” parasite, ie parasites can hide in cell, divide, multiply then burst
  • Survive & divide within cells of the reticuloendothelial system, e.g. macrophages of the liver, spleen, bone marrow and skin
    Amastigotes reside with a phagolysosomal vacuole a.k.a. parasitophorous vacuole, not free in the cytoplasm

Amastigotes survive and multiply within the macrophage by modifying innate pathogen-killing mechanisms such as Inhibiting the generation of free radicals and down-regulating IL-12 and suppressing Th1 cell activation. Promoting the production of immunosuppressive molecules

In sand fly vector = flagellated promastigote

706
Q

Diagnostic techniques for leishmaniasis?

A

Parasitology -> Microscopy (+/- culture)
Eg LN puncture (lowest sensitivity), Splenic (best sensitivity) then BM aspiration, lesion biopsy for CL

Immunodiagnosis
IFAT/ELISA
DAT (direct agglutination test)
rK39 Ag-based (ELISA and dipstick format) -> now a dipstick and almost universally used as part of diagnostic algorithm for VL
Katex (Ag detection)

DNA probes and PCR
Kinetoplast DNA (minicircle)
16-18s rDNA gene- species specific

707
Q

Summary of diagnostic techniques for visceral leishmaniasis?

A

Parasitology
- Near 100% specificity
- Only way to diagnose non-response/relapse
Splenic aspirate is the most sensitive
- Diagnose primary VL (if borderline DAT)
- Assess post-treatment efficacy
- Diagnose VL relapse

rK39 RDT
- South Asia - All formats perform well
- East Africa – level of sensitivity is insufficient to rule out VL; sensitivity sufficient if case-definition is respected; best tool for decentralised diagnosis

DAT
- More sensitive that rK39 RDTs
- Should be used in pts with a –ve rK39 RDT where clinical suspicion of VL is still high

VL+HIV patients: rK39 + DAT combination, plus biopsy

East African setting: rK39 + biopsy

708
Q

Two key causes of visceral leishmaniasis?

A

L.donovani or L.infantum

709
Q

What is the vector for leishmaniasis? Can you give some examples of species?

A
  • Phlebotomus spp. - Europe, Africa, S. Asia
  • Lutzomyia spp. - South America

Unique sand fly – Leishmania species pairings
Ph. sergenti & L. tropica; Ph. papatasi & L. major L. longipalpis & L. infantum

710
Q

Zoonotic cause of visceral leishmaniasis?

A

L.infantum

711
Q

Zoonotic cause of cutenous leishmaniasis?

A

L.mexicana
L.major

712
Q

Anthroponotic causes of cutenous leishmaniasis?

A

L.tropica

713
Q

Features and risk factors of mucocutaneous leishmaniasis?

A
  • Reappearance of previously resolved L.Viannia sub-genus:
    ○ L.V.braziliensis (1-10%)
    Others also possible: L. V.panamensis, L.V.peruviana, L.V.guyanenis months to years
  • Initial skin lesion that may spontaneously heal –> mucosal involvement months to years later
    Eg ulceration, destruction of nasal and oral tissues, disfigurement

Risk factors:
○ Male>female, age, nutritional status, # of LCL lesions above waist, inadequate Rx, immune status

  • ~10% will develop MCL, trigger unknown
  • Tissues of soft palate are infected, decrease amastigotes
    Can be called immune-mediated inflammatory reaction -> Th1 cells producing IFN-γ (interferon-gamma), which helps activate macrophages to kill the parasites but causes tissue damage disproportionate to tissue load

Destructive and difficult to treat

714
Q

Describe the spectrum of disease in Leishmaniasis with respect to immunity and briefly the role of the immune system in disease control vs progression

A

Leishmaniasis disease works on spectrum
- Hypersensitivity
○ MCL
○ DL
○ ~LCL, LR
○ Th1 cells release IFN-Y, greater response than Th2 cells releasing IL-10
○ Means immune-system mediated damage is large component of pathology
- Moderate: PKDL
- Hyposensitivity
○ ADCL
Greater Th2 response + release of IL-10 than Th1 response with release of IFN-Y

Disease control occurs due to
- Ag presentation and activation of macrophages
- Proinflammatory cytokines and chemokine response
- Ab production
- Decrease in lesion size and parasite no.
- ROS and NO production
- Cure is associated with reduction in IL-10 and IL-13 in VL and increased IFN-Y and CD4 T cell response in CL

Disease progression associated with
- Reduced Ag presentation
- Increased IL-10 and TGF-beta
- Non-productive Ab, increase in lesion size and no. of parasites
- ROS inhibition

715
Q

How can measles impact the eye?

A

Acutely: Conjunctivitis

  • Vitamin A deficiency induced by measles
    ○ Drying of cornea
    ○ Keratomalacia
    ○ Corneal ulcers and ulcerative keratitis
    ○ Optic neuritis
716
Q

Bilateral rash on palms?

A

syphilis until proven otherwise

secondary

717
Q

Reasons for high STI prevalence in LMICs?

A
  • Lack of access to affordable healthcare and effective treatment
  • Demographic factors eg lots of young people in Africa + no. of partners
  • Rural to urban migration
  • Migrant labour
  • Commercial sex
  • Different patterns of sexual networks (eg younger women with older men)
718
Q

Equation and influencing factors to consider transmission of STIs? For each of these factors what interventions can be enacted to reduce transmission of STIs?

A

R0 = beta x c x D

R0: basic reproductive number
beta: probability of STI transmission given exposure
c: Exposure rate of susceptible to infectious partners (eg rate of sexual partner change)
D: duration of infectious period (eg HIV is v long)
________________________________________________
Efficiency of transmission (β):
– infectivity of the organisms (viral or bacterial load)
– biological interactions with other organisms (HIV & STIs)
– sexual behaviours which increase or decrease risk (condom use, anal vs vaginal vs oral sex vs non penetrative)

Behavioural parameters (c):
– rate of partner change
– patterns of sexual mixing (gender, age, assortative/disassortative mixing)

Duration of infection (D):
– infectious period of the organism
– treatment-seeking behaviour
________________________________________________
On β (=reducing efficiency of transmission)
- Treat
- treat cofactors (e.g. treat STIs to reduce HIV shedding)
- condoms & safer sex practices
- male circumcision
On c (=reducing the rate of exposure)
- rate of partner change
- contact with high-risk partners
- concurrent partnerships
On D (=reducing the duration of infectiousness)
- implement prompt STI diagnosis and effective case management
- promote early treatment-seeking (through education / IEC)

719
Q

What is a cornerstone of STI case management?

A

Partner notification and management

Clinical goal: to provide concurrent antibiotic treatment to the sexual partner(s) to eradicate the STI and prevent reinfection

Public health goal: to interrupt the chain of transmission and reduce spread

Little evidence of effectiveness in LMIC & potential harmful effects (violence, incomplete treatments)

720
Q

Prevention methods for STIs

A

1 Partner notification

2 Female condoms

3 Microbicides

4 Voluntary medical male circumcision (VMMC)

721
Q

Diagnosis and treatment methods for STIs (ie 3 theoretical methods)?

A

Clinical aetiological diagnosis:
Clinical features, clinical acumen, reproducibility, self-
medication

Laboratory aetiological diagnosis:
Diagnose specific pathogens via multiple methods

Syndromic approach:
Consistent group of symptoms and clinical signs => target ALL treatable pathogens that can cause the syndrome (ie give treatment for all possible causes)

722
Q

Recommendation for WHO for STIs were testing isn’t available?

A

Syndromic approach, 3 syndromes:
- Urethral Discharge & Cervical Discharge
- Vaginal Discharge
- Genital Ulcer

Treat for all possible causes of whichever syndrome you treat with

Diagnosis + treatment +:
+ the “4 Cs”:
Compliance
Counselling
Condoms
Contacts

723
Q

Urethritis and cervical discharge
Causative organisms of this syndrome:

A
  • Neisseira gonorrhoeae
  • Chlamydia trachomatis
  • Trichomonas vaginalis
  • Mycoplasma genitalium
724
Q

Diagnosis and treatment of urethritis and cervical discharge syndrome?

A

If you can do molecular assays and can get result back on same day then do that
Otherwise treat for chlamydia and gonorrhoea
+ offer test for HIV and syphilis

  • Gram stain is rapid but only 50% sensitive for Ng in women
  • Culture is the reference standard for Ng but requires a microbiology laboratory
  • Near patient lateral flows & microscopy available for Trichomonas
  • NAATs may be used to diagnose all infections using self-administered vaginal swabs

Treatment:
GC: Ceftriaxone 250mg AND Azithromycin 1gm

CT: Doxycycline 100mg BD 14/7 OR Azithromycin 1gm (NB Only if not given azithromycin for GC)

TV: Metronidzole 2gm OD – OR – 400mg BD for 7 days

MG: Complicated & only routinely done in a limited number of high-
income countries (Azithromycin 1st line)

725
Q

Vaginal discharge
Causative organisms of syndrome:
Diagnosis?
Treatment?

A

Trichomonas vaginalis
Candida albicans
Bacterial vaginosis
Chlamydia trachomatis
Neisseira gonorrhoeae

DIAGNOSIS
Molecular assay often not available
No good lateral flow for chlamydia
Could maybe do speculum
Otherwise:
- Treat for NG + CT
- Consider treating for BV/TV if abnormal vaginal discharge or micro/molecular results
Consider treating for candida if symptomatic

TREATMENT
Candidiasis (thrush): Clotrimazole pessary STAT – or - Miconazole pessary - or - Fluconazole 150mg p.o. STAT

TV: Metronidazole 2.0g STAT or 400mg BD for 7 days

BV: Metronidazole 400 or 500mg bd daily for 7 days

726
Q

Causative organisms of genital ulcer disease? Diagnostic pathway?

A

Herpes Simplex Virus
Treponema pallidum (Syphilis)
Haemophilus ducreyi (Chancroid)
Klebsiella granulomatis (Donavanosis)
Lymphogranuloma venereum

If ulcer vesicular or hx of recurrent ulcers in same site -> treat for HSV, if not then treat for HSV and syphilis
If NO hx of recent syphilis treatment (past 3 months) -> treat for syphilis

Diagnosis:
Ideally have molecular assay eg NAAT, if not
* Clinical diagnosis is unreliable
* Dark field microscopy is effective for primary syphilis but
requires a microbiology laboratory
* Culture is possible (but difficult) for chancroid
* NAATs may be used to diagnose all infections

727
Q

HSV key facts and treatment?

A

Herpes simplex virus type 2 or type 1
- Lifelong infection with clinical recurrences
- Symptoms are self-limiting
- Symptoms more severe and of longer duration if immuno-compromised

Treatment: Acyclovir 400mg tds for 5-10d
- Not necessary in most cases
- Suppressive treatment can prevent recurrences
- Long term Rx may be needed in immuno-compromised patients

728
Q

Chancroid key facts and treatment?

A

Haemophilus ducreyi
Multiple painful ulcers with regional lymphadenopathy
Mainly seen in core groups (i.e FSW) -> hard to maintain outside of this as lesions are painful so people seek treatment
Asymptomatic infection rare

Treatment = Azithromycin

729
Q

Lymphogranuloma venereum (LGV) key facts, presentation and treatment?

A

Cause: Chlamydia trachomatis, L1,2, 3

Clinical features:
Primary: Small, transient genital ulcer or proctitis

Secondary:
- Lymphadenopathy -> (inguinal or retroperitoneal), usually unilateral
- Continuing proctitis
- Fever, rigors, weight loss, erythema nodosum

Tertiary: Fibrosis, lymphoedema, rectal stricture

Treatment: Azithromycin OR Doxycycline (better)

730
Q

Donovanosis (Granuloma inguinale) key facts, presentation and treatment?

A

Klebsiella (formerly Calymmatobacterium) granulomatis

Possibly still prevalent in India, PNG, Brazil, Guyanas

Clinical features:
* Beefy red genital/anal ulcers, usually painless
* Inguinal lesions – bubo, ulcer
* Lymphoedema, fibrosis, autoamputation of penis

Treatment: Azithromycin

731
Q

Complications of STIs for women?

A
  • PID
    ○ Endometritis
    ○ Salpingitis
    ○ Peritonitis, perihepatitis
  • Infertility
  • Ectopic pregnancy
732
Q

Baby develops gonococcal disease during birth, what is the name of this syndrome? How do you 1) treat and 2) prevent it?

A

Gonococcal opthalmia neonatorum

Treatment:
CLEAN EYES IMMEDIATELY after delivery
- Single dose IM ceftriaxone 125mg is highly
Alternatives
- Kanamycin 25mg/kg IM stat (max 75mg)
OR
- Spectinomycin 25mg/kg IM stat (max 75mg)

Prevention:
* 1% tetracycline ointment is as good as 1% silver nitrate
* 2.5% povidone-iodine is more effective than 1% silver nitrate or
* 0.5% erythromycin

NB: ophthalmia neonatorum -> can lead to corneal ulceration and blindness in 24 hours (remember to also screen for Chlymydia)

733
Q

What is the relationship between STIs and HIV? What studies examined this relationship and did they work?

A
  1. Increased Susceptibility to HIV:
    Biological factors: inflammation + disruption to mucosal barrier in the genital and rectal tracts due to STIs -> entry points for HIV -> easier for the virus to infect cells

Immune response: STIs recruitment of immune cells, including CD4+ T cells (which HIV targets), to the site of infection -> increases the concentration of target cells for HIV, facilitating its transmission

  1. Increased Transmission of HIV:
    Higher viral load: STI can increase HIV viral load in genital secretions -> more infectious to others
    Ulcerative STIs: STIs that cause open sores eg syphilis or herpes -> direct access for the HIV virus to enter or leave the body -> amplifying the risk of transmission.
  2. Co-infection and Disease Progression:
    STIs can accelerate HIV disease progression by continuously activating the immune system, which HIV exploits to replicate. Persistent STIs can lead to a faster depletion of CD4+ T cells, advancing the course of HIV more rapidly.

Can we treat STIs to reduce HIV?
Three major RCTs were conducted in 1990s-early 2000s
* Mwanza – Syndromic STD management
- Reduced rate of new HIV within those communities
* Rakai– Mass STD treatment every 10 months
- Did not work
* Masaka– Information, education & communication plus syndromic STD management
- Did not work
Differences - ?reflect different stages of HIV epidemic and different burdens of STIs in population

734
Q

Serological tests for syphilis?

A

Treponemal
* Lab and POCT
* Highly specific
* Stay positive for life

Non-treponemal
* VDRL & RPR (rapid plasma reagin test)
* Less specific
* Titres rise & fall with treatment

735
Q

Treatment of syphilis?

A

Primary, secondary and early latent
– Benzathine penicillin G 2.4 million units IM stat

Late latent
– Benzathine penicillin G 2.4 million units IM weekly x3

No RCT evidence underpinning different treatment for Late Latent

736
Q

How does mother to child transmission occur for syphilis?

A

Transmission believed to occur in-utero

Risk varies by stage
– Primary 60%
– Secondary 90%
– Early 40%
– Late Latent <10%
Associated with spirochetaemia

Most vertical transmission is in women with latent syphilis -> as most women are in this stage

737
Q

Paediatric syphilis complications?

A
  • 30-40% stillbirth, fetal demise, hydrops fetalis or preterm birth if mother untreated
  • In inadequately/untreated mothers: meningitis, snuffles, hepatosplenomegaly, osteochondritis, periostitis, rash, haemolytic anaemia, thrombocytopenia

> 2 years old, ie late symptoms: developmental delay, sensorineural hearing loss, interstitial keratitis, hutchinson’s teeth, saber skins, clutton joints

738
Q

Elimination of congenital syphilis: targets
recommended by WHO?

A
  • > 95% of antenatal clinic attenders screened
  • > 95% of seropositive women treated
  • Incidence of congenital syphilis <50 per 100,000 births (including stillbirths)
739
Q

What can we use as a proxy for risk in Syphilis?

A

Serology eg high RPR titre more likely to represent active infection

More associated with still birth and poor birth outcomes

740
Q

What is the benefit for on site rapid testing for syphilis?

A
  • Proved to be most cost effective

○ Ie despite the fact it will remain positive for life, even if a person comes back and receives repeat treatment its cheaper to treat than to prove they actually have a repeat infection

Dual tests are too expensive

741
Q

WHO recommendation for treatment of syphilis?

A

Early Syphilis: 1 dose IM Benzathine Benzylpenicillin
Late or UNKNOWN: 3 weekly doses IM Benzathine Benzylpenicillin

Issues:
* Most women don’t know duration = recommend 3 doses
* Good completion for first dose in many settings BUT poor completion all 3 doses
* Some data suggests this might be ok -> ie 1 dose is enough to prevent all the v severe outcomes like stillbirth/LBW etc but still don’t know whether this would actually be the case for women who have latent/long infection

742
Q

Treatment recommendations for infants with syphilis

A

Complex algorithms depending on if infant is “High” or “Low” risk

High Risk:
- Infants with signs of congenital syphilis
- Positive body fluid / placenta by Darkfield of PCR
- Infant RPR 4* or higher than mother
- RX: Procaine Penicillin or Benzylpenicillin for 10 Days

Medium Risk:
- The mother was ‘inadequately treated’
- RX: Procaine Penicillin or Benzylpenicillin for 10 Days OR IM Benzathine

Low Risk:
- The mother was ‘adequately treated’
Rx: Nil or Single Dose Benzathine Penicillin

743
Q

Pros and cons of centralised lab testing compared to point of care testing?

A

Centralised laboratory testing
* High throughput, highly sensitive and specific PCR platforms (+)
* Multiplex capabilities (+)
* Robust laboratory system/network required (-)

Point-of-care testing
* Reduced time to treatment (+)
* All activities complete on same day (+)
* Supply chain complexity (-)
* Quality assurance more difficult to monitor (-)

744
Q

Key criteria that should be met for rapid point of care test?

A
  • Real time connectivity
  • Ease of specimen collection
  • Affordable
  • Sensitive
  • Specific
  • User-friendly
  • Rapid & Robust
  • Equipment-free and Environmentally friendly
  • Deliverable to end-users
745
Q

STIs that have strong associations with adverse birth outcomes?

A
  • Syphilis
    ○ >50% of pregnancies in women with syphilis will result in adverse birth outcome
  • Trichomoniasis
  • Chlamydia
  • Gonorrhoea
746
Q

IPAZ study - what and what did it show?

A

Strategy for integration of POC diagnostics for STIs into antenatal care in Zimbabwe. Only 8% of ANC attendees could be approached for enrolment but did find STI in 31% of asymptomatic pregnant women (most common CT), 98% of which were treated on same day. Issues with study provision in terms of environment, getting staff involved, device limitations and client willing to wait.

747
Q

Wantaim study - what and what did it show?

A

Wantaim study
- Aimed to see if POC testing and immediate treatment of CT, Gon., Trich. Or BV in pregnancy ?lead to reduction in preterm birth +/- LBW compared with standard antenatal care
- Big study (n>4000), Papua new guinea
- No sig difference between two arms
- No impact among women who had chlamydia or trichomonas at enrolment
- 39% had CT/Gon./Trich at enrolment in intervention arm
CT prevalence in control group dropped -> ?due to malaria prophylaxis standard of care which can have some effect in treating CT

?Jury is out as to impact of POC testing for STIs on birth outcomes -> new trials planned in Zim and Botswana

748
Q

What are two key components of social innervation to increase STI uptake?

A

Social innovation:
a community-engaged process that sees the wisdom of communities and impacts social and health outcomes

Prosocial interventions:
one example of social innovation that encourage voluntary actions to help others

-> One prosocial intervention is pay-it-forward which provides a financial nudge (zero cost health service) and a social nudge (handwritten postcard) to increase uptake of services

749
Q

2030 WHO targets for cervical cancer elimination?

A
  • 90% of girls fully vaccinated with HPV vaccine by 15 years of age
  • 70% of women are screened with high-performance test by 35 and 45 years of age
  • 90% of women identified with cervical disease (precancer or cancer) received treatment and care
750
Q

HPV
- Type of virus?
- Key proteins?
- Key genotypes for invasive disease?
- Genotypes linked to anogenital warts?

How common is the infection?
What is associated with persistence?

A

HPV - double-stranded DNA within a spherical shell (capsid), composed of
- structural proteins L1 and L2
* current vaccines target L1
- Viral proteins E1, E2, E4, E5, E6, E7

40 genotypes infect the genital area, 15 high-risk (HR) oncogenic types,
=> 2 types (HPV16/18) linked to 70% of invasive cervical cancer
=> 7 types (HPV16/18 + 31/33/45/52/58) linked to >90% of invasive cervical cancer

HPV6 & 11 linked to anogenital warts (AGW)

HPV is the most common sexually transmitted infection, 50-80% of women will acquire HPV in their lifetime
- Most infections will clear; median duration of new infection is approx. 8 month
- Although new infections decrease with age, risk of persistence increases with age
- HR-HPV persistence increases risk of precancerous lesions and cancer

751
Q

What is the Ab response of
- HPV infection
- HPV vaccination?

A

~ 50% of women develop no measurable antibody response following HPV infection

In women who have detectable antibody levels following natural infection, levels of antibodies are low (compared to post vaccination levels)

Low levels of antibodies are less likely to protect against re-infection

752
Q

Relationship of HPV and HIV?

A
  • HPV is RF for HIV acquisition (x2)
  • HIV increases HPV incidence (x2) and decreased HPV clearance
  • Increase multiple HR infection and broader range
  • HIV increases cervical cancer (x6)
  • HIV increases anal cancer in MSM (x10) and women (x7-10)
  • Increase AGW prevalence, incidence and persistence

Impact of ART?
- Women on ART have 17% reduced risk of HR-HPV prevalence
- Reduction in SIL incidence and progression
- Decrease in ICC incidence
- Risk is lower if initiated early (higher nadir CD4)
+ prolonged ART duration, good adherence & effective treatment etc

753
Q

What tools can prevent cervical cancer?

A

· Primary prevention -> HPV vaccine

· Secondary prevention -> HPV DNA, visual inspection, cytology “pap” smear

· Treatment precancer -> ablation/excision

· Treatment cancer -> surgery, radio, chemo

754
Q

What is the impact of screening programmes on invasive cervical cancer?

A

Decreasing in HIC, Europe/North America, South America and Oceania, Asia:
* Successful screening and treatment programmes
* HPV vaccination
* Changes in disease risk factors

Increasing in Eastern European and Sub-Saharan Africa:
* Shorter duration and quality of screening
programmes
* Fewer vaccination programmes
* Changes in disease risk factors
* Impact of HIV

755
Q

WHO recommendations for cervical cancer prevention?

A

General population of women:
Screen and Treat OR Screen, Triage and Treat * HPV DNA as primary screening test
* Starting at age 30
* Every 5 to 10 years screening interval

For women living with HIV:
Screen, Triage and Treat
* HPV DNA as primary screening test
* Starting at age 25
* Every 3 to 5 years screening interval

  • Where HPV DNA testing is not yet operational, use a regular screening interval of
    every 3 years when using VIA or cytology as the primary screening test among WLHIV
  • Use either provider or self-collected samples
756
Q

Issue with using HPV DNA as screening test for women living with HIV?

A

Low specificity for high grade lesions

757
Q

What HPV vaccines exist?

A

Virus-like particle (VLP) vaccines
* 2-valent: 16/18 (GSK Biologicals; Innovax)
* 4-valent: 6/11 + 16/18
* 9-valent: 6/11 + 16/18 + 31/33/52/58 & 45

Use 4/9 in gender non-discriminative settings to help prevent ano-genital warts

Prevent HPV acquisition, persistence
and neoplasia development
(vaccines are prophylactic, NOT therapeutic)

  • Type specific protection++
  • Some cross protection (eg. 31/33/52 & 45)

Vaccines MOST effective when introduced PRIOR to exposure
Ie prior to sexual debut

> 80% effective in preventing invasive cervical cancer

758
Q

What is HPV vaccine coverage like globally? What are some issues with it?

A

141 countries have HPV vaccine in national programme

  • 40% LMICs vs 88% HIC
  • 1st dose coverage greater in LMICs than HIV but lower for 2nd dose -> drop out

53% (75/141) of country HPV vaccine programs were gender neutral

Challenges:
Costs of vaccine and costs of delivery
Political will & support
Competing priorities
Perception of cervical cancer prevention as a public health priority
Vaccine hesitancy
Appropriate delivery strategies
Vaccine supplies
COVID-19

759
Q

What is the WHO recommendation (since 2022) to increase HPV vaccine coverage?

A

Option: 1-dose schedule for 9 to 20-year-olds

Prioritize the vaccination of Immunocompromised/HIV+
populations – also at ages beyond primary target - with at least2 doses, ideally 3

760
Q

Examples of parasites causing CNS infection

A
  • Toxoplasma gondii
  • Neurocysticercosis/ Taenia solium (Pork tapeworm larva FigureD)
  • Trypanosoma brucei spp. (African trypanosomiasis/Sleeping sickness)
  • Plasmodium falciparum (Cerebral malaria)
761
Q

Viral causes of CNS infection

A

Herpes simplex virus
Varicella zoster virus
Enteroviruses
HIV
Arboviruses: Japanese encephalitis virus, Dengue virus, Chikungunya virus
Rabies virus
Herpes B virus
Nipah virus

762
Q

Bacterial causes of CNS infection

A

Strep pneumoniae
Neisseria meningitidis
Haemophilus influenzae
Listeria monocytogenes (Young/Old/Immunosuppressed/Pregnant)
Mycobacterium tuberculosis
Mycoplasma pneumoniae
Streptococcus suis
Burkholderia pseudomallei
Orientia tsutsugamushi (Scrub typhus) and Rickettsia spp.
Spirochaetes: Treponema pallidum, Borrelia spp.
(Lyme, Relapsing fever)
Bacterial Toxins

763
Q

Differential diagnoses for JE?

A
  • Dengue virus infection
  • Rickettsial disease ( esp. Scrub typhus)
  • Enterovirus infection
  • Chikungunya virus infection
  • Leptospirosis
  • Cryptococcal meningoencephalitis
764
Q

How to diagnose JE?

A

Gold standard: Seroneutralisation
- Rise in anti-JEV Iggs as measured by haemagglutination inhibition

Issues with serology due to cross-reactivity

CSF or serum IgM antibody detection (by ELISA) is the primary diagnostic method
- MAC ELISA, problems with specificity
- Often can’t get CSF then relying on serum sample -> reduces specificity even further

PCR tests may also be used, especially early in infection

765
Q

What is the WHO case definition for JEV?

A
  1. JEV IgM antibody in cerebrospinal fluid (CSF) or serum, as detected by
    anti-JEV IgM-capture ELISA (MAC-ELISA)
  2. Detection of JEV antigens in tissue by immunohistochemistry
  3. JEV genome in serum, plasma, blood, CSF or tissue by reverse
    transcriptase polymerase chain reaction or an equally sensitive and specific
    nucleic acid amplification test
  4. x4/> rise in anti-JEV Igs as measured by haemagglutination inhibition (HI)
    or plaque reduction neutralization assay (PRNT) in serum collected during
    the acute and convalescent phase (>14 days later) and tests performed in
    parallel with other confirmatory tests to prevent cross-reactivity.

NB: MAC ELISA is WHO recommended reference standard. This is the test used in
endemic countries. Problem: needs lab, time, skilled workers, CSF, etc Has poor field sensitivity and reduced specificity when using serum so ideally need CSF.

766
Q

Types of vaccine for JE? How long should immunity last for?

A

4 main types: inactivated MB, inactivated VC, live-attenuated, Chimeric
- Inactivated JE vaccine (e.g., JE-VAX) for adults and children over 1 year
- Inactivated MB: 2 doses in endemic areas or 3 doses non-endemic + booster, concern regarding potential link with ADEM and reduced supply
- Inactivated VC: one used in UK

  • Live attenuated vaccine (e.g., SA 14-14-2) used in some endemic areas; provides longer-lasting immunity
  • JE-YF chimeric vaccine

Immunity should be life long

767
Q

Key facts regarding JE?

A
  • JEV is an arbovirus (Culex sp.) and a flavivirus (like dengue, zika, yellow fever, west nile virus…)
  • Chief identified cause of acute CNS infections in Asia
  • JE mainly occurs in rural areas; Humans are accidental hosts; 1/300 develop clinical disease; mainly children affected in endemic
    areas
  • The clinical presentation is typically acute encephalitis syndrome (fever, headache, reduced GCS, seizures, movement disorders)
  • No treatment but JEV vaccine recommended by WHO and supported by GAVI as part of childhood immunisation programme in endemic countries
  • 24 endemic countries; 15 are now vaccinating, but limits in diagnostic capacity affect our true understanding of the burden and evaluate vaccination, and there are ongoing problems with vaccine coverage
  • JEV is a zoonosis and vaccination will not eradicate disease
768
Q

Definition of arbovirus?

A

(short for arthropod-borne viruses) - broad group of viruses transmitted by arthropods such as mosquitoes, ticks, and sandflies

Includes:
- flaviviruses eg Dengue, Zika, West Nile, yellow fever
- Togaviridae eg Chikungunya
- Bunyaviridae eg rift valley fever

769
Q

Key facts JE on
- Causative agent, transmission, Endemic regions, incubation, clinical presentation

A

Causative Agent

Caused by Japanese encephalitis virus (JEV), a flavivirus related to West Nile, Zika, and dengue viruses

Transmission

Primarily transmitted by Culex mosquitoes (especially Culex tritaeniorhynchus), which breed in rice paddies and standing water
- Pigs = amplifying hosts
- Human/horses = dead-end reservoir

Endemic Regions

Found mainly in Asia and parts of the Western Pacific. Countries with significant risk include India, Nepal, Thailand, and Vietnam

Incubation Period

Typically 5-15 days after the mosquito bite

Symptoms

Many infections are asymptomatic. Symptomatic cases often have:
- Initial symptoms: Fever, headache, vomiting.
- Neurological signs: Seizures, altered mental status, tremors, stiffness, which can lead to coma.
- Severe cases may progress to brain inflammation and long-term neurological damage.

Mortality Rate

Case fatality rate is approximately 20-30% in symptomatic cases; up to 50% of survivors have permanent neurological or psychiatric issues

770
Q

Pathogenesis of rabies

A
  1. Infection via: broken skin, intact mucous membrane, tissue transplant
  2. Incubation, usually 20-90 days (can be weeks to years)
  3. Virus enters nerve endings, evades immune surveillance, centripetal retrograde axonal transport to brain
  4. CNS infection: transsynaptic spread, viral replication, inclusion (Negri) body formation, neurons intact but dysfunctional
  5. Clinical signs of encephalomyelitis begin +/- immune response
    Centrifugal neuronal transport to salivary glands (viral excretion), virus carried to skin, heart, muscle, tongue eg but no viraemia
771
Q

Main cause of Rabies in South America since dogs have been controlled?

Other types of rabies transmission?

A

Vampire bat

99% of human transmission is from dogs
Can come from other animals eg seals, bats, racoons
human to human -> organ transplantation from donors who died of unknown neurological illness

772
Q

Differential diagnosis paralytic rabies

A

*Post-vaccinal encephalomyelitis
*Paralytic poliomyelitis
*Flavivirus myelitis
*Herpes simiae (B virus) encephalomyelitis

So provide full Rx, until diagnosis of rabies made

773
Q

Prodromal symptom of furious and paralytic rabies?

A

Itchiness - ? due to rabies virus reaching dorsal root ganglion

774
Q

Symptoms of furious vs paralytic rabies

A

Furious -> die quickly
* Phases of arousal and lucid intervals
* Cranial nerve lesions III,VII, VIII
* Autonomic stimulation: secretions, temp control
* Cardiac arrhythmias
* Coma
* Paralysis
* Priapism
* Axonal neuropathy

Paralytic -> die slowly
* Ascending paralysis, loss of tendon
reflexes
* Fasciculations
* Sphincter dysfunction
* Fever, sweating, gooseflesh
* Bulbar / respiratory paralysis
* (hydrophobia)
* Survive < 30 days

775
Q

Key feature of furious rabies?

A

Hydrophobia

  • Provoked by drinking / draught of air (aerophobia)
  • Reflex forceful jerky inspiratory muscle spasms
  • With inexplicable terror
  • Violent inspiratory gasps- contraction of
    diaphragm
  • Generalized extensor response
    → opisthotonos, convulsion, cardiac/ respir.
    arrest
    → pneumothorax, oesophageal tears
  • Later on - provoked by minor stimuli

? due to virus wanting to remain in head and neck nerves and avoid GI distribution etc

776
Q

Complications of Rabies?

A

Heart: arrhythmias, hypotension, cardiac failure

Lungs: asphyxiation, pneumonia, pneumothorax respiratory arrhythmias: inspiratory spasms periodic breathing- cluster breathing, Cheyne-Stokes ARDS, respiratory failure

CNS: Convulsions, hypo-/ hyper-pyrexia, diabetes insipidus, inappropriate ADH secretion

Gut: Bleeding, stress ulceration

777
Q

Does rabies cause meningitis?

A

NO

only encephalitis syndrome

778
Q

Diagnostic Ix for rabies?

A

Skin biopsy (NUCAL BIOPSY FOR HAIR FOLLICLES) = GOLD STANDARD
* Immunofluorescence: Ag (skin punch biopsy)
* RT-PCR
* Detect Ag detection and viral RNA detection

Saliva, tears, CSF (need multiple, 2-3 a day, sequential)
- Tissue culture, mouse innoculation
- RT-PCR
- Virus isolation

Serum, CSF
- Neutralising Ab
- Serology

779
Q

A sample from an encephalopathic patient shows negri bodies on microscopy from cerebellum. What is the diagnosis?

A

Rabies

Bodies in cytoplasm of neuronal cells

780
Q

When should you attempt ICU care to aim at survival for rabies patient?

A

Attempt ICU only if
1) American bat virus implicated
2) Early appearance of Ab
3) ITU facilities available

781
Q

Rabies pre-exposure vaccination - what are the benefits and who should be vaccinated?

A

Benefits
- No need for post-exposure Rabies Immunoglobulin
- Decreases number of post-exposure vaccinations required
- Helps in cases where post-exposure treatment is delayed
- Might provide protection against inapparent rabies exposure cases

Who
* Those who live in highly endemic areas
* Rabies virus laboratory workers*
* Those who handle animals in/from endemic areas eg. dog catchers, wildlife workers, zoos, ports.**
* Travellers to, or residents of, dog rabies enzootic areas, especially children.
* Hospital staff likely to care for rabies patients – Very low risk but overcomes fear

*need antibodies checked (>0.5IU/L) every 1-2 years and booster if low
** need antibodies checked (>0.5IU/L) every 2 years and booster if low

782
Q

Pre-exposure vaccination options for Rabies? What is the advised dosing schedule?

A
  • IM (deltoid)
    ○ 1 dose = 1 vial
    ○ If delayed vaccination should be resumed, not restarted
    ○ A change in route is acceptable if unavoidable -> restarting course is not necessary
    ○ Vaccine cold chain required 2-8 degrees
    ○ Standard regimen in high resource countries BUT too expensive for most LMICs
  • ID
    ○ Improves availability, accessibility and affordability
    ○ More immunogenic because dermal dendritic cells convey vaccine Ag to reticulo-endothelial system
    ○ 1 dose =0.1 mL (irrespective of WHO recommended vaccine brand)
    ○ Store opened vial up to 6 h
    ○ Site: deltoids, thighs or suprascapular
    ○ Appropriately trained health care staff essential
    ○ Vaccine failure if given IM/SC instead

WHO say 2 dose schedule on DAY 0 and 7: but in UK and other countries 3rd dose is given as WHO could provide no evidence as to immunogenicity without 3rd dose

783
Q

WHO advised dosing schedule for pre-exposure vaccination for rabies?

A

2 dose schedule on DAY 0 and 7: Rabies vaccine*: 1 dose intramuscular (IM)
or intradermal (ID) 2 sites at each visit

but in UK and other countries 3 dose is given as WHO could provide no evidence as to immunogenicity from 3 doses, 3rd dose given at 28 days

In immunocompromised patients ideally check neutralising Ab checked at 2-4 weeks if feasible

784
Q

Management of immunocompromised person with possible rabies exposure?

A

Post-exposure prophylaxis (PEP):
* Meticulous wound cleaning
* Irrespective of their rabies vaccination status:
* 5 doses of vaccine: days 0, 3, 7, 14 and 28-30
AND
* RIG for all Category 2 and 3
cases

785
Q

Do you give RIG to a CAT 3 rabies exposure if they are vaccinated before?

A

NO
If they have been vaccinated then even if cat 2 or 3 exposure they still don’t need RIG

just
- wash wound with soap for 15mins
- vaccinate

786
Q

Prevention methods for rabies?

A
  • Mass dog vaccination/contraception
    programs.
  • Educate children to avoid mammal interaction
  • Vigorous washing of all bite wounds immediately with soap
  • Human vaccination: Rabies vaccines are outstandingly effective, no one should die
    of dog-transmitted infection
787
Q

Need to know facts on Rabies

A
  • Rabies is caused by zoonotic, neurotropic Lyssa viruses transmitted through saliva of an infected mammals. Incubation period 2-3 months (range days to years)
  • Dog rabies accounts for 99% human infections worldwide.
  • The combination of pre- and post-exposure vaccination has proved 100% successful in preventing human infection
  • Rabies is an acute, progressive viral encephalomyelitis that is almost universally fatal. Virus is already widely disseminated by the time of onset of clinical symptoms.
  • Pre-exposure prophylaxis means you do not need post-exposure rabies immunoglobulin (passive immunisation) (unless immunocompromised)
  • Intensive care treatment may be appropriate if infected by a bat in the Americas or if antibody is present. Otherwise, compassionate palliative care is the best management of rabies patients
  • Control of canine rabies is the most economical method of preventing human rabies.
788
Q

Chagas
- What
- Vector
- Transmission
- Symptoms
- Treatment
- Key trials

A

Trypanosoma cruzi
Transmitted by inoculation of contaminated faeces of a triatomine vector

Transmission:
- Vector
- Vertical (transplacental)
- Transfusion
- Transplantation
- Oral ingestion
○ On the rise
Eg juice like acai juice, higher burden of parasite and higher mortality rates

Vectors:
- Triatoma infesters, Rhodnius prolix, Triatoma dimidiata

Acute:
- Self-limiting syndrome 4-8 weeks
- Fever, malaise, lymphadenopathy, heptosplenomegaly, myocarditis
- Chagoma may occur at entry site
- Rare case presenting as orbital cellulitis (eye acts as another portal of entry) = Romana’s sign
Rarely, meningoencephalitis

Indeterminate (2/3), Chronic/determinate (1/3)
- Arrhythmias (BBB, SN disruption, AV block)
- Myocardial abnormalities (segmental wall motion abnormalities, dilated cardiomyopathy)
- Aneurysms (LV, RV)
- Thromboembolism (stroke>systemic)

Treatment
- Benznidazole (or nifurtimox)
- Named patient order to WHO outside of endemic areas
- 60 days -> poorly tolerated
Objectives
- Effective for parasitological clearance
- Effect upon clinical endpoints unproven
- Prevents vertical transmission

TRIALS
- BENEFIT -> did not show evidence in patients with early cardiomyopathy that treatment with Benznidazole did not affect progression but patients did clear parasites (?relevance)

BENDITA trial
- Looked at benznidazole alone and in combination with fosravuconazole
- Similar rates of seroconversion in BZN shorter dose in 2 weeks compared to 8 weeks
- So should we be treating for shorter courses than 60 days

MULTIBENZ
- No differences in sustained negativity by qPCR between control with Benz, low-dose and short treatment
- Can we shorten treatment to 2 weeks
Marked low response to treatment in Brazil site -> not explained by genotype

789
Q

What are the two forms of Chagas in immunosuppressed people?

A
  • HIV -> SOL (mimics cerebral toxo), meningocencephalitis (ring enhancing lesions)
  • Transplant recipients -> fever, rash, myocarditis
790
Q

A man presents with left periorbital edema and erythema. Conjunctival infection. He recently took a trip for 6 months to rural Bolivia. What is the diagnosis?

A

Romana’s sign

Chagas

791
Q

Diagnosis of Chagas?

A

Serology (need 2x tests)
- ELISA
- IFAT
- In indeterminate phase titre is unhelpful, response to therapy variable -> ie can’t use as marker of disease or therapy

PCR - Parasite DNA detection
- Only performed if sero-positive
- May be positive in indeterminate phase
- Conversion seen with therapy but not necessarily sustained neither reflective of preventing disease progression
- Used for congenital infection

Microscopy (blood/buffy coat) only useful in acute infection (<1% diagnosed) and congenital (50% sensitivity)

792
Q

Key trials for Chagas disease?

A

TRIALS
- BENEFIT -> did not show evidence in patients with early cardiomyopathy that treatment with Benznidazole did not affect progression but patients did clear parasites (?relevance)

BENDITA trial
- Looked at benznidazole alone and in combination with fosravuconazole
- Similar rates of seroconversion in BZN shorter dose in 2 weeks compared to 8 weeks
- So should we be treating for shorter courses than 60 days

MULTIBENZ
- No differences in sustained negativity by qPCR between control with Benz, low-dose and short treatment
- Can we shorten treatment to 2 weeks
Marked low response to treatment in Brazil site -> not explained by genotype

793
Q

Should you treat a pregnant patient with Chagas to prevent vertical transmission?

A

NO - treat after breast feeding has stopped

  1. vertical transmission occurs in 5% of pregnancies in women with T. cruzi infection
  2. without maternal testing this transmission is invisible until the development of disease in young adult life
  3. anti-parasitic treatment of infants is highly effective so early detection and treatment is critical
  4. anti-parasitic treatment during pregnancy is contra-indicated, but future pregnancies can be protected by treatment of mother after breast-feeding ceases
  5. pre-conception anti-parasitic treatment prevents almost all vertical transmission
794
Q

Recommended strategy to precent vertical transmission in non-endemic countries

A

Girls and women from endemic countries of Latin America should be offered testing (only needed once)
1. Offer screening (ideally pre-conception) to all women of child-bearing age
2. Offer antenatal screening to all women from Latin America
3. Test and follow-up infants born to T. cruzi seropositive mothers, treating those with confirmed infection
Babies will have positive serology from mum so need to take PCR

794
Q

What is the transmission of VHF? What drives outbreaks?

A

· Zoonotic spillover to humans : activities of infected animals/vectors and humans overlap

· Unpredictable combination of factors:
○ shedding patterns,
○ land use change, social/economic conditions,
○ wrong place wrong time

· human-vector interface: animal husbandry, household environment

· Exception ‘human reservoirs’ : where viable virus persists in survivors after recovery, e.g. EV sexual transmission

· Person to person transmission drives outbreaks
○ R0: estimates from 1.34 to 4.71 but limited data

○ Direct contact with contaminated body fluids or objects through broken skin or mucous membranes
○ High potential for exposure in households due to caring practices, and late presentation to healthcare due to fear
○ Super-spreading events: funeral ceremonies, markets, cultural obligations
○ Increased population movement: shift into more dense urban environments changes exposure risk
○ Nosocomial transmission due to poor IPC, undetected cases, slow diagnosis of suspect cases

*Exception
Ongoing exposure to a common zoonotic transmitter – can resemble person to person spread (e.g. Lassa)

795
Q

Who is at risk of VHFs?

A

People in contact with potential transmitting vector eg animal faeces, insects etc

Household members and close contacts

Nosocomial infection

796
Q

Describe Asymptomatic, pauci-symptomatic, and symptomatic states in differing proportions in the context of VHF

A

Asymptomatic
− Ebola 2.6-6.5%; CCHF ~70%; Lassa
80%; Marburg 0%?
− Difficulty of identifying due to non-
specific common symptoms and case
definition
− Unlikely to transmit due to lack of
body fluids & low viral load

Pauci-symptomatic
− Transmission potential may depend
on type of symptoms (“dry”/ “wet”)
− Increasing viral load linked to onset of
more florid symptoms

Symptomatic
− extent of exposure to a symptomatic case,
and whether that case is dry or wet is
strongly associated with risk of infection
− Ebola - many contacts with high exposure
do not become infected.

797
Q

What are some of the key principles of outbreak management?

A

Reduce exposure, break transmission, speed care for infected, reduce severity & fatality
__
Active case detection : reduce delay from onset
to isolation, speed care provision

Earlier and more rapid isolation & care: improve
outcome, reduce exposure

Triage & suspect management: avert
undetected cases in routine services; also
confidence in safety in healthcare setting

Faster, more frequent testing: reduce time to
care, better management of suspect & quarantine

IPC healthcare facilities: prevent transmission,
avert amplification

IPC household: human and animal: prevent/
reduce exposure

“IPC” community: adapt public health measures
- avert exposure

Contact tracing: early detection, break
transmission

Quarantine: reduce time to detection, reduce
unwitting transmission but risk of opposite effect

798
Q

What are the different species of orthoebolavirus genus and which ones are associated with outbreaks in humans?

A

Genus orthoebolavirus has 6 different species associated with it (only top 3 associated with :
- Zaire/Ebolavirus (highest mortality rate)
- Sudan ebolavirus virus
- Bundibugyo ebolavirus
- Tai forest ebolavirus
- Reston ebolavirus
- Bombali ebolavirus

799
Q

Pathophysiology of Ebola?

A
  • Targets mononuclear phagocyte system -> affects dendritic cells (inhibition of RIG-I and aberrant maturation) and macrophages
  • Rapidly replicates in the cytoplasm + disseminates to nearby tissues
  • Immune system dysregulation -> B and T cell dysregulation
  • Cytokine storm
  • Vascular leakage, third spacing and coagulopathy (haemorrhage), damage to endothelium, low BP
  • Liver: transaminitis, reduced clotting factor production, hypoalbuminaemia, hypoglycaemia
  • GI: V&D, fluid and electrolyte loss, disrupted acid-base balance, hypoproteinaemia
  • Spleen: cytokine dysregulation and death of lymphocytes
  • Kidney: AKI, oliguria, anuria, electrolyte abnormalities
  • Skeletal muscle: injury + elevated CK
800
Q

Ebola (Zidare) summary
- Epi & Transmission
- Clinical presentation
- Diagnosis
- Treatment
- Prevention

A

Cycle of transmission
- Single pass from animal to human eg via bat faeces etc
- Amplification in humans, human to human transmission

Epi

Major outbreaks include the 2014-2016 West Africa outbreak and periodic outbreaks in the DRC.
- Guinea, Liberia, Sierra Leone
High case fatality rate (often 50% or higher), with outcomes heavily impacted by healthcare access.
Ebola’s severe impact on public health emphasizes the need for rapid response, strong healthcare measures, and international collaboration.

Transmission

Animal-to-Human: Likely spread from infected wild animals, such as fruit bats, through direct contact or consumption
Human-to-Human: Direct contact with bodily fluids (blood, saliva, sweat, etc.) or contaminated surfaces and materials
Non-Airborne: Ebola does not spread through air, water, or casual contact

Pathogenesis

  • Targets mononuclear phagocyte system -> affects dendritic cells (inhibition of RIG-I and aberrant maturation) and macrophages
  • Rapidly replicates in the cytoplasm + disseminates to nearby tissues
  • Immune system dysregulation -> B and T cell dysregulation
  • Cytokine storm
  • Vascular leakage, third spacing and coagulopathy (haemorrhage), damage to endothelium, low BP

Clinical presentation
Incubation Period: 2-21 days
- Only symptomatic patients can spread the disease and remain infectious until virus is not in blood
Early phase: day 0-1 fever, muscle pain, headache, sore throat, fatigue (non-specific, eg could look like Malaria)
Day 2-4: some diarrhoea, hiccups, feel worse etc

Progressive Symptoms (GI phase): day 4-9: Vomiting, diarrhoea (can be bloody), abdominal pain, rash, kidney/liver impairment
Cardiovascular compromise, weak/fast pulses, less ambulation of patients
Different phenotypes e.g. renal, encephalopathy, hypoglycaemia

Terminal phase: Internal/external bleeding, organ failure
Day 10-12, confusion and delirium worsen, oliguric, anuric, death (both gradual and sudden)

For those who survive, they may still be shedding -> transmission risk

Lab diagnosis
Gold standard: RT-PCR
Other:
RDTs (used as initial test, often need PCR to confirm):
- OraQuick Ebola
ELISA
Viral culture (rarely used), high biosafety risk)

Other:
- Elevated transaminases
- Raised CRP tracked with fevers
- Raised WCC and thrombocytopenia

Treatment

Supportive Care:
- IVF aggressively
- Electrolyte abnormalities correction
- Septic shock physiology -> IVF, monitor for third spacing
- Symptomatic mx
- Prophylactic Abx use due to concerns about bacterial gut translocation
- Nutrition etc
Experimental Treatments: Antivirals and monoclonal antibodies
- Inmazeb = REGN-EB3 = 3 Ab targeting Ebola glycoproteins to get entry
- Ebanga = mAb114 = single Ab targeting - more effective it administered early
Vaccine: rVSV-ZEBOV vaccine (licensed 2019) protects against Zaire Ebola strain, given in outbreak zones.

Prevention

PPE: Essential for healthcare workers and those handling deceased.
Safe Burial Practices: Reduces transmission from deceased individuals.
Public Health Measures: Quarantine, isolation, contact tracing help contain outbreaks.
Vaccine Deployment: Targeted in high-risk areas to prevent spread.

Case management

Shift from isolation unit to Ebola treatment unit
- Trace
- Isolate
- Treat

801
Q

In Uganda in Sudan virus outbreak of 2022 what experimental therapeutics were used compassionately?

A

Remdesevir

MBP134 (monoclonal Ab targeting SUDV glycoprotein)

802
Q

Epidemiology case definitions for Lassa?

A
  1. Suspected case
    Any individual presenting with one or more of the following:
    Malaise, fever, headache, sore throat, cough, nausea, vomiting, diarrhoea,
    myalgia, chest pain, Hearing loss, and either
    a. History of contact with excreta or urine of rodents
    b. History of contact with a probable or confirmed Lassa fever case within a period of 21 days of onset of symptoms OR Any person with inexplicable bleeding/hemorrhage.
  2. Confirmed case
    A suspected case with laboratory confirmation (positive IgM Antibody, PCR or virus isolation)
  3. Probable case
    A suspected case (see definition above) who died or absconded without collection of specimen for laboratory testing
803
Q

Lassa fever key facts
- Epi and transmission
- Clinical presentation
- Diagnosis
- Treatment and prevention

A

Virus
Arenaviridae family
Lassa virus
RNA virus with high mutation rates

Transmission
Most due to contact with rodent excreta (urine, feces, blood) from infected Mastomys natalensis rats
- Ingestion of contaminated food
- Handling, killing, ingesting rodents
- Inhalation of aersolized virus (eg during sweeping or cleaning surfaces)
Human-to-human transmission can occur via direct contact with bodily fluids, sexual transmission (persistence up to 12 months)
Nosocomial
Vertical + breastmilk

Endemic Areas
Found mainly in West Africa, especially in Nigeria, Sierra Leone, Liberia, Guinea, and Benin
Some seasonal trends
Highest in Nigeria, outbreaks Dec-May
Some imported cases -> fatality higher due to delays in diagnosis (secondary transmission from these v rare)
Approximately 100,000–300,000 cases per year

Incubation Period
Typically 1-3 weeks after exposure Asymptomatic

Early symptoms:
Fever, malaise, sore throat, sorethroat, myalgia (similar to Malaria)
Severe symptoms:
Bleeding, facial swelling, shock, and multi-organ failure
Complications:
Renal, neurological (seizures, encephalitis), bleeding, resp, pregnancy (termination, abortions, eclampsia)

Mortality Rate
Generally 1% for all cases
Up to 15–20% in severe hospitalized cases
Higher risk of death in late-stage pregnancy (up to 80%)

Diagnosis
RT-PCR on blood samples, sometimes on CSF from children
ELISA for antigen or antibody detection
Virus isolation in specialized labs

RDT being evaluated
Treatment
Ribavirin (antiviral) is effective if given early
Supportive care: IVF, renal support, blood products, resp support

Prevention
No licensed vaccine currently available
Prevention relies on rodent control, proper food storage, and protective measures for healthcare workers (e.g., PPE)

Public Health Impact
Significant impact on healthcare systems in endemic regions; outbreaks pose high risks due to human-to-human transmission in healthcare settings

804
Q

CCHF roadmap aims and timings?

A

Vision
* To be able to reduce death and morbidity from CCHF through safe and
affordable effective treatments informed by rapid, reliable, simple-to-use and
easily accessible diagnostics by 2023 (missed)

and
* To be able to prevent or mitigate CCHF disease through deployment of safe,
affordable and effective vaccines or other preventive measures by 2030

805
Q

Summary of CCHF
Transmission
Epi
Clinical features
Diagnosis
Management
PH approach

A

Crimean-Congo Hemorrhagic Fever virus (CCHFV)
Nairovirus genus in the Bunyaviridae family
RNA virus
Priority pathogen by WHO 2014

Transmission
Tick-borne zoonosis -> Hyalomma ticks
Maintained in nature -> enzoonotic tick-vertebrate eg camels/cows-tick cycles
Primary human infection: via tick bite OR direct contact with blood or tissues of infected ticks or viraemic vertebrates including wild animals and livestock
- High risk due to unsafe slaughter of animals
Secondary human to human transmission: direct contact with blood, bodily fluids/tissues of infected (not extensive, hospital worse spread)

High transmission risk when providing direct patient care or handling dead
Concern transfer in migratory birds into Spain/Europe (carry ticks with them)

Epidemiology
Found in parts of Africa, Asia, Eastern Europe, and the Middle East - Turkey, Iraq, AFG, Iran, Pakistan
Areas with high tick populations, especially rural and farming regions, are at higher risk.

Incubation Period
Typically 1-13 days, 3-4 days time to presentation/hospital admission, hospitalisation for 8 days, median age 50 years
Typically 1-3 after tick bite
Depends on transmission route (shorter with tick bites, longer with exposure to fluids)

Symptoms
Classified by fever, thrombocytopenia, haemorrhage -> VHF that ‘bleeds’
Children: - Fever, anorexia, N&V, Abdo pain -> not death, milder disease
Initial symptoms:
- Fever (not that high), headache, muscle aches, nausea, and abdominal pain
- Transaminitis and bleeding, very low platelets BUT patient initially may have NEWs0/1 and look otherwise well, apart from thrombocytopenia and bleeding (different to ebolavirus)
Progressive symptoms can include severe bleeding, shock, and organ failure

Prognostic indicators + scoring criteria
Fatal outcome associated with:
- Elevated APTT, ALT, LDH, Plts <20
Criterias:
- Swanepoel
- SGS scores
- SSI parameters
Mortality Rate
Varies between 10–40%, with higher rates in untreated severe cases.

Diagnosis
RT-PCR (blood, v little from urine)
- Development of portable real-time PCR
ELISA for CCHFV antigen or antibody detection
Virus isolation in specialized laboratories (high risk)

RDT: CCHF prototype 001 in development
Gn Mab: BH14, JE12

Treatment
Supportive care
Ribavirin: Cochrane review showed low quality evidence, some benefit
Favipiravir
mAb development -> 2 selected with broad-neutralising activity (being taken from recovered patients and also lab based)

Prevention
One health approach essential
No licensed vaccine is available
- Anti-tick and Anti-CCHFV (DNA vaccines) showing promising results

Prevention
Relies on tick control measures, use of protective clothing, insect repellents, and avoiding animal contact in endemic areas

Public Health Impact
High impact in endemic areas; CCHF can cause outbreaks in healthcare settings due to person-to-person transmission. Special precautions are needed in hospitals to prevent spread

806
Q

Life cycle ecchinococcus

A
  • Eggs in Environment: The life cycle starts when adult Echinococcus tapeworms residing in the intestines of definitive hosts release eggs into the environment through the host’s feces.
    • Ingestion by Intermediate Hosts: Intermediate hosts, including livestock (sheep, cattle, pigs) or accidental human hosts, ingest these eggs via contaminated food, water, or soil.
    • Larval Development in Intermediate Host: Once ingested, the eggs hatch in the intestines, releasing oncospheres(larvae) that penetrate the intestinal wall and migrate through the bloodstream to various organs, primarily the liver and lungs. Here, they develop into cysts (hydatid cysts in E. granulosus or alveolar cysts in E. multilocularis), which grow slowly and produce infectious protoscolices (immature tapeworms).
    • Definitive Host Infection: Carnivores become infected when they consume the organs of intermediate hosts containing these cysts. The protoscolices attach to the intestinal lining of the definitive host, mature into adult tapeworms, and complete the cycle by producing more eggs.
    • Human Infection: Humans act as accidental intermediate hosts. If they ingest Echinococcus eggs, the larvae form cysts, primarily in the liver and lungs, causing cystic or alveolar echinococcosis, which can be life-threatening if untreated.
807
Q

Differentials for cyst in echinococcosis?

A
  • Cystic lesion in the liver
  • Simple cyst
  • Haematoma
  • Abscess
  • Cystadenoma
  • Cystadenocarcinoma
  • Necrotic liver mets
808
Q

Which Echinococcus granulosis related cysts need treatment?

A

Viable cysts
Transitional cysts

DO NOT need to treat non-viable cysts

809
Q

Management of Echinococcus granulosis cysts?

A

Depends on 1) Type, 2) Size, 3) Position
Includes:
- Conservative
- Medical: albendazole, praziquantel
- Interventional: PAIR, PEVAC
- Surgical

General principles
· Albendazole alone: cysts <5cm
· PAIR + ABZ: cysts >5cm without daughter cysts or solid areas
· Surgery
○ Cysts > 5cm with daughter cysts or solid areas
○ Complex cysts invading other structures or the biliary tree
○ Lung cysts

810
Q

How do you make a diagnosis of Cysticercosis?

A

The diagnosis of NCC should be based on neuroimaging and supported by specific serological tests. Assays of choice are LLGP-EITB for antibodies and mAb-based ELISA for antigen detection

Imaging -> ideally both CT (for calcifications) or MRI (better image resolution and for intraventricular cysts) to detect cysts
- Establish diagnosis
- Define characteristics of infection

Serological tests (e.g., Western blot, ELISA) for antibodies
- Small amounts of Ag produced by parasite
- Western blot almost 100% sensitive with 2 or more parasites
- Not good for follow up
- For Ag may be helpful in follow up

Molecular
- PCR on serum, CSF, urine
Works in EP NCC but sensitivity unknown in PNCC

811
Q

Pro vs cons of treating neurocysticercosis?

A

Pros:
Rapid disappearance of cysts.
* Severe cases are seen less frequently now.
* Series of ABZ or PZQ-treated patients have
better clinical evolution (have less seizures) than
not-treated patients seen at the same centers.
* Less residual calcifications.(?)

Cons:
NCC becomes symptomatic after several years,
when the parasite dies.
Anti-parasitic treatment causes severe, acute,
unnecessary brain inflammation.
Treatment-induced inflammation or stroke can kill the patient.

812
Q

Neurocysticercosis
- Cause
- Epi
- Life cycle
- Transmission
- Types
- Symptoms

A

Caused by Taenia solium (pork tapeworm)
Humans ingest tapeworm eggs from contaminated food, water, or direct contact with infected individuals

Epi
South America, Sub-Saharan Africa, South East Asia, also North Asia - Major cause of neurological disease

Life Cycle
1. Ingestion of infected pork, poorly cooked Taeniasis -> faecal contamination
2. Eggs ingested by humans develop into larvae (cysticerci) that migrate through the bloodstream
3. Larvae encyst in tissues, especially muscles and the brain
4. Simultaenous ingestion of T.Solium eggs or proglottids -> porcine cysticercosis

Transmission
Fecal-oral route through contact with infected feces or contaminated food/water
Autoinfection (self-infection) can occur if a person ingests tapeworm eggs from their own intestines

Types
Cysticercosis: Larval cysts in various body tissues, such as muscles, skin, eye
Neurocysticercosis: Larval cysts in the brain, causing neurological symptoms
- Intraparenchymal: cyst -> granuloma -> calcification (or disappearance)
○ Variable evolution
○ Other: massive infections
- Extra-parenchymal:
○ Subarachnoid “racemose” cysts
○ Not quite associated to seizures and epilepsy: intracranial HTN, hydrocephalus
○ Significant mortality
○ Uncontrolled growth of cystic membrane

Symptoms
Cysticercosis: Often asymptomatic but may cause lumps under the skin or muscle pain
Neurocysticercosis: Seizures (PNCC), headaches, intracranial HTN (NPNCC)
- Can present as any neurological system as can get cysts anywhere in brain

813
Q

Neurocysticercosis management, prevention and PH impact

A

Treatment
Antiparasitic drugs (e.g., albendazole, praziquantel) to kill larvae
- In general beneficial, need to identify type of CC, prioritise symptomatic management, its never an emergency (ie don’t give acutely)
- BUT in some people causes extensive brain inflammation -> stroke/inflammation can kill patient
- BUT seems to reduce seizure incidence
Corticosteroids to reduce inflammation in neurocysticercosis
- Indication and dose not yet certain
Antiepileptic drugs for seizure management.
Surgery may be required for certain cases

Prevention
Proper cooking of pork to kill tapeworms
Improved sanitation and hand hygiene to prevent egg ingestion
Education on safe food practices and avoiding contaminated water or food sources

Public Health Impact
Significant in regions with poor sanitation and where pork is commonly consumed, such as Latin America, sub-Saharan Africa, and parts of Asia
Leading cause of acquired epilepsy in endemic areas

814
Q

What is leptospirosis caused by? Where is it found, how is it transmitted and what is the pathophysiology of the disease?

A

Leptospira spp. = spirochaetes
Free living and aerobic, motile
Divided into serogroups based on serology not on DNA

Transmission: ZOONOSIS
Proximal tubules of mammals (primary reservoir) -> direct or indirect contact with animal urine
Rodents are most important for maintaining transmission
Survives for weeks in fresh water
Spirochaetes invade skin and mucus membranes

RFs:
Occupational or recreational exposure to contaminated water/soil, cuts/abrasions, inadequate footwear, farming, sewerage workers

Epi:
Tropical and temperate regions, worse in rainy season, men>women, 1 million cases, 3rd commonest infectious cause of life threatening disease in returning travellers

815
Q

Pathophysiology of leptospirosis?

A

Pathophysiology:
Inoculation -> haematogenous spread and dissemination –> sepis syndrome/cytokine storm (IL6 and IL10)
Severity related to innoculum dose and host factors
Organ damage from direct pathogen insult and host response
Haemorrhage can occur -> endothelial damage, capillary fragility and thrombocytopenia

816
Q

Clinical s/s of leptospirosis? Complications:

A

Incubation period: 7-14 days, can range from 1-30

BIPHASIC (but manifested in few people in reality)

(Most can be mild, self limiting illness)

Early phase: leptospiraemia -> non-specific febrile illness lasting 3-9 days, fever chills myalgia OF CALVES, headache, 1/2 patients D&V, 1/3 cough

Possible brief period of defervescence but two phases can overlap

Immunological phase: IgM in blood, leptospira in urine, complications in 10%

WEIL’s disease in 5-10%: Fever, Jaundice, AKI, Conjunctival suffusion (develops in first week of illness)

Complications:
○ Rhabdomyolysis
○ Jaundice
○ Acute Kidney Injury (with hypokalaemia)
○ Pulmonary haemorrhage (>50% CFR)
○ Myocarditis
○ Aseptic meningitis
○ Multi-organ failure
○ Rarer:
▪ Encephalitis
▪ Peripheral Nervous System: neuritis/radiculitis/optic neuritis

Risk of Jarisch-Heixheimer reaction in 20% of patients

817
Q

Features of lab findings and diagnostic techniques for leptospirosis?

A

Laboratory findings:
* Non-specific:
○ Leucocytosis, thrombocytopenia
○ Elevated CRP/Procalcitonin
○ AKI (with hypokalaemia)
○ Elevated conjugated bilirubin (+-transaminitis and ALP rise)
○ +- anaemia
○ NB clotting normal, no DIC
Urinalysis: protein/blood/pyuria

Diagnosis:
* Compatible clinical syndrome
* PLUS - Exposure history
* PLUS - Detection of leptospira DNA (PCR, blood 1st week, urine 1st week onwards, high specificity, ok sens.) or serological response (IgM) (ELISA) (cross reactivity issue, ok spec/sens)

Other diagnostic techniques
○ Dark field microscopy (need at least 10 spirochaetes)

○ Culture of blood (1st week illness, prolonged incubation of 12 weeks and low diagnostic yield)

○ Microscopic agglutination test (MAT) -> serological gold standard

Caution with clinical risk/predictor scores e.g. modified Faine’s -> Poor specificity and poor PPV

818
Q

Management of leptospirosis?

A
  • OPD vs Inpatient
  • Majority mild and spontaneously resolve. Standard IPC precautions.
  • Antibiotics – consider EARLY:
    ○ B-lactams/cephalosporins: IV Benzyl-penicillin or Ceftriaxone
    Doxycycline PO (or azithromycin PO or Amoxicillin PO)

Cochrane review showed insufficient evidence.

Supportive management:
○ Hydrate, correct electrolyte disturbances
○ Early haemo/peritoneal dialysis
○ Ventilation (low tidal volumes, “stiff lungs”)
○ Transfusion
○ Cardiac monitoring
○ No specific evidence based therapy

RISK: Jarisch-Herxheimer reaction

819
Q

What is the Jarisch-Herxheimer reaction?

A

Systemic reaction

Occurs hours after initial treatment of spirochete infections, such as syphilis, leptospirosis, Lyme disease, and relapsing fever

Presents with fever, rigors, hypotension, and flushing

Can easily be mistaken for a drug-induced hypersensitivity reaction

The underlying mechanism is initiated by antibiotic-induced release of spirochete-derived pyrogens

Transient rises in TNF, IL-6, and IL-8 have been detected

Reaction lasts 12–24 hours and can be alleviated by aspirin/ibuprofen/prednisone

820
Q

Features of CNS leptospirosis?

A
  • Lymphocytic pleocytosis (moderate)
  • Elevated protein, normal glucose
  • Imaging:
    ○ Cortical, midbrain, and rarely subcortical white matter change
  • Can mimic viral meningoencephalitis
  • Prognosis:
    ○ Generally good
    ○ Can have long term sequelae in severe cases
821
Q

Prevention techniques for leptospirosis?

A

Sanitation

Modifying risks and related behaviours -> provide wellies in wet season

Abx prophylaxis: 200mg doxy weekly (debatable)

Vacccination
10-15years away for humans
Routine for animals - tetravalent vaccine

822
Q

Most common presentation of melioidosis in children?

A

Parotid abscesses

823
Q

Melioidosis
- Cause & RFs
- Epi
- Transmission

A

Burkholderia pseudomallei
Gram negative, oxidase positive -> environmental saprophyte
- Seasonal, lives in soil -> rain -> increased exposure -> affects rural poor
- Long latency (highest reported 28 years)
Opportunistic pathogen so need predisposing conditions:
- Diabetes mellitus *high risk
- ETOH
- Renal failure
- Steroids
- Exposure to ++ innoculum

Epi:
- Endemic in 45 countries
- Found south of equator, issue in SE Asia and Australia (eg Darwin)
- Thailand has surveillance programme

Transmission:
- Innoculation
- Inhalation

824
Q

Melioidosis
- Clinical presentation

A

Variable clinical presentation -> “Sepsis, pneumonia, abscesses anywhere in body”

In order of prevalence:
1) Lung: pneumonia, pul abscesses
2) CVS: bacteraemia, pericarditis, aneurysm
3) GI: liver/splenic abscesses (honeycomb appearance)
4) UT: acute pyelo, kidney/prostatic abscess
5) Skin/soft tissue: ulceration, soft tissue abscess
6) Head/neck: parotid abscess (think kids), neck abscess, lymphadenitis
7) MSK: septic arthritis, osteomyelitis, myositis
8) CNS: encephalomyelitis, brain abscess

Other eg mastitis, mediastinal mass, corneal ulceration, epididymo-orchiotis, scrotal abscess

825
Q

Melioidosis
- Diagnosis
- Management

A

Staining: safety pin polarity (not exclusive to this pathogen though, also seen in Yersinia pestis etc)

Culture = gold standard
○ Est. sensitivity 60%
○ Enhanced if right samples from correctly suspected patients: ie blood, pus if abscess, urine etc

Serology
○ Problems with sensitivity (particularly with indirect agglutination test) and specificity
○ Standardisation

PCR
○ Not routinely used anywhere

Ag detection -> research attempting for RDT

826
Q

Gram negative bug that stains with safety pin polarity, what is it?

A

Burkholderia pseudomallei

827
Q

Concern regarding burkholderia pseudomallei

A

Hazard group 3/Tier 1 ‘select agent’ -> risk of bioterrorism

For
* Availability
* Intrinsic resistance
* Environmental persistence
* High mortality
* Inhalation transmission
* Wide host range

Against
* Little development of weaponisation
* No person to person spread
* Less of effect in immunocompetent (inoculum dependent)
* Difficult to disseminate (eg not like anthrax in forming spores)

828
Q

What is a major issue with treating melioidosis?

A
  • Intrinsic resistance to early beta-lactams and aminoglycosides
  • Resistance to co-trimox overcalled by disc diffusion
  • Acquired resistance to beta-lactams can occur
    ○ Uncommon
    ○ Rarely necessitates change in tx
  • No person to person spread
  • People get sick from environment not from other people so reduces AMR risk/burden
829
Q

Treatment for melioidosis?

A
  • Intensive phase: 2+ weeks IV beta-lactam (ceftazidime or carbapenem)
    • Eradication phase: 12+ weeks PO Co-trimox
      ○ Essentially all manifestations get 2 weeks minimum of intensive phase management and then depending on manifestations, varying length of treatment for both intensive and then for eradication phase
      ○ Eg CNS infection can get total treatment course of 3+ months
      Supportive management: critical to managing the illness
830
Q

Plague
- Cause & RFs
- Epi
- Transmission

A

Yersinia pestis, gram negative bacteria
3 main forms:
- Bubonic
- Septicaemic
- Pneumonic

Circulates in animal reservoirs -> rodents

Epi:
Broad-belt across tropics
Large burden (>90%) in Africa, eg DRC, Madagascar, Uganda, Tanzania
More sporadic in Asian countries -> less contact with reservoir
2017 outbreak in Madagascar

Transmission:
* Droplet
* Direct physical contact eg touching, sexual, unsafe burial
* Indirect contact eg touching contaminated soil or surface
* Airborne
* Faecal-oral
* Vector borne - carried by insects eg fleas from infected animals

831
Q

Cycle of transmission for Yersinia pestis

A

○ In humans generally occurred by flea bite which was infected by biting rodent carrying the disease

○ Bacteria can multiply inside the flea -> stick together to form biofilm plug -> blocks stomach -> causes it to starve

○ Flea bites host -> continues trying to feed even though it can’t quell its hunger -> flea vomits blood tainted with bacteria back into bite wound

○ Bacterium then infects a new victim -> flea dies from starvation

○ Rats die first –> in time then plague outbreak

Urban cycle: refers to infected fleas biting rodents, then humans coming into contact with them, or new fleas being infected by them

Sylvatic cycle: infected flea infects wild rodent which can then infect humans to again fleas can become infected

832
Q

Pathogenesis of plague?

A
  • Survival and proliferation within macrophages
  • Array of toxins and type 3 secretion mechanism: ~2 extra plasmids
    ○ pMT1: carries ymt -> for biofilm
    ○ pPCP1: carries pla -> for bubos/invasion at tissue barriers
  • Molecular typing being used in epidemics
833
Q

Three types of plague and relevant symptoms?

A
  • Buboes
    ○ Bubo=a tender, enlarged and inflamed lymph node) enlarged smooth tender/painful lymph nodes related to the site of a flea bite or other exposure
    ○ BUBONIC PLAGUE
  • Septicaemia with multiorgan failure/DIC -> peripheral gangrene: SEPTICAEMIC PLAGUE
    ○ Bacteraemia + role of endotoxins
    ○ N & V & Abdo pain
    ○ Hypotension + organ failure + DIC + localised areas of tissue/organ death (?black)
    ○ May be 2nday to bubonic/pneumonic or could be primary presentation
    ○ High mortality
  • A rapidly progressive pneumonia: PNEUMONIC PLAGUE
    ○ Rapidly progressive pneumonia
    ○ Short incubation - 2-4 days
    ○ Fever + weakness + nausea
    ○ Then pleuritic pain, SOB, cough, blood tinged sputum (contains numerous bacilli)
    ○ CXR: severe pneumonia +/- mediastinal or hilar lymphadenopathy
    ○ High mortality
  • Others
    ○ e.g meningeal involvement following dissemination
    ○ Pharyngeal and cellulocutaneous kinds

All characterised by invasive bacterial infection and then uncontrolled proliferation of Y.pestis

834
Q

Common presentation of yersinia pestis infection?

A
  • Short incubation period: 1-7 days
  • Bubo forms
    ○ Often singular
    ○ Common in groin
    ○ May occur in axilla/cervical region related to site of initial bite/scratch
    ○ May not be fluctuant or contain pus
  • Pain may be first feature - ie before swelling appears
  • Progressive -> rapid onset of sepsis features and dissemination:
    ○ Chills
    ○ Malaise
    ○ High fever eg 39
    ○ Muscle cramps
    ○ Seizures
    ○ Peripheral gangrene secondary to sepsis
835
Q

What antibiotic class does NOT work for Yersinia pestis infection?

A

Macrolides

836
Q

WHO treatment recommendations for Yersinia pestis
- pneumonic, septicaemic or bubonic plague?
- Meningitis
- Post exposure presumptive treatment
- Pre exposure prophylaxis

A

Pneumonic, septicaemia, bubonic
In general:
Fluoroquinolones (ciprofloxacin, levofloxacin and moxifloxacin) added to the first-line
medicines (streptomycin and gentamicin)
e.g.
Streptomycin or gentamicin
Fluoroquinolones 10 days
For bubonic -> doxy also

Meningitis
In general: Floro. with good CNS penetration
Moxifloxacin
Ofloxacin
Chloramphenicol at least 10 days

Post-exposure:
Floro added to first line of doxy and co-trimox.
e.g.
Ciprofloxacin
Doxycycline
Co-trimoxazole 7 days

Pre-exposure:
Tetracycline
Doxycycline
Co-trimoxazole

837
Q

Investigations for diagnosis of Yersinia pestis?

A
  • LN aspirate
    ○ Culture organisms -> often bubos don’t contain pus so can instil small vol of saline and withdraw it again
  • BCs
    ○ Can see organisms in blood smears if patient is septicaemic
    ○ In early course of illness: smears may be negative via microscopy but culture should be positive
  • Sputum
    ○ Culture possible from sputum of v ill pneumonic patients
    ○ Blood usually positive at that time
  • Bronchial/tracheal washings
    ○ Can take from pneumonic patients but other bacteria can mask presence of plague
  • Post-mortem
    ○ Take lymphoid, spleen, lung, liver or BM via DFA (direct florescent Abx) or PCR
  • RAPID tets
    ○ Lateral flow
    § F1RDT detects F1 capsular antigen (think that makes biofilm and blocks up stomach of flea) of Y.pestis which is present in a large amount of buboes, blood and sputum from infected patients
    § Performed on sputum from patients with suspected pneumonai plague and on bubo aspirate from patients with suspected bubonic plague
    Easily interpreted/used
838
Q

Hallmark features of Yersinia pestis on culture?

A

Growth at 22-25oC

An irregular or “fried egg” appearance, at 48-72 hours

NB: Gram stain shows safety pin appearance (like Burkholderia pseudomallei)

839
Q

Prevention methods for Yersinia pestis?
Control of cases?

A
  • Reduce likelihood of infected flea bites
  • Reduce direct contact with infectious tissues and exudates
  • Reduce resp exposure to pneumonia plague cases
  • Pest control
  • Flea control
  • Wear gloves when handling wildlife
  • Plague vaccine unreliable, worked for bubonic but not pneumonic, (but new developments)

CONTROL
* Report
* Isolate and treat clothing and baggage to rid of fleas
* Disinfect
○ Surfaces contaminated with sputum
○ Flea control in environment
* Quarantine in effective
○ Close contact of pneumonic cases -> chemoprophylaxis (tetracyclines or chloramphenicol) and surveillance for 7 days
* Investigation of contacts
* Specific treatments

840
Q

Actions to manage an epidemic of plague?

A

Investigate all cases if possible with autopsy and
lab tests

Communicate to avoid panic

Intensive flea control with insecticidal dusts

Bodies, clothing, habitats

(Reduce/attack rat populations)

Contact tracing and prophylaxis

Protect field workers (powder and repellents)

Possibility of deliberate release an issue (most
cases pneumonic? Yp lives in the air for an hour)

841
Q

How does yersinia pseudotuberculosis manifest?

A

a self limiting enterocolitis associated with mesenteric adenitis, ileitis and gut wall granuloma formation

842
Q

Three relevant plasmids required for Yersinia pathogenesis?

A

pCD1
- encodes type 3 secretion system, covers bac in needle like projections which inject yersinia outer proteins (yops) into host cells
- yops induce apoptosis, inhibit phagocytosis and cytokine production and leukocyte recruitment

pMT1
- produces an antiphagocytic slime layer around the bacterium which prevents phagocytosis + blocks flea stomach

NB: this is the target of the RDT
Ymt, which is required for survival within the midgut of the flea and is therefore important for
the ability of Y. pestis to be transmitted by fleas

pPCP
lasminogen activator protease Pla which is thought to promote bacterial dissemination at the inoculation site by inhibiting the formation of a fibrin clot barrier, inhibiting innate immune cell
recruitment, and inactivating antimicrobial peptides

843
Q

Features of Zika virus:
- Virus
- Transmission cycles
- Epi

A

Small enveloped single-positive stranded RNA virus = FLAVIVIRUS
Two genotypes: African and Asian

Africa:
- Enzootic sylvatic transmission cycle, between non-human primates and forest dwelling Aedes mosquitoes with epidemic spillover into humans in peri-urban/urban areas via Aedes
- Also human-to-human transmission: sexually, vertically, transplanted blood or tissue

Cycle not identified yet in Asia but likely exists

844
Q

Where are most Zika viruses in the UK now from?

A

Used to be central/South America during epidemic but NOW

from south and SE Asia

845
Q

Clinical presentation of Zika?

A

50-80%: asymptomatic

Symptomatic:
Incubation 2-14 days
Rash (maculopapular), diffuse, palms and soles
+/- pruritis
Low grade fever (often self-reported)
Arthralgia
Conjunctivitis (can help differentiate from dengue)

Normally self-limiting illness, 2-7 days, with full recovery

NB: prior Zika virus may worsen dengue disease caused by serotypes 2, 3 and 4, but conversely prior dengue infection may protect against zika

846
Q

How should diagnosis of zika be made?

A

PCR
Single positive PCR (blood or urine) -> test both
- blood often only briefly positive within first week of illness

Negative PCR does not rule out illness

Serology
For Zika-specific IgM and IgG -> available + diagnostically useful
- Prone to cross-reacting with other flaviviruses SO caution when interpreting

NB: IgM for Zika may not appear in patients with a history of previous dengue infection, an example of ‘original antigenic sin’

Negative serology 2 weeks after the onset of symptoms makes the diagnosis of Zika highly unlikely.

847
Q

Describe the neurological consequences of Zika infection

A

Zika virus demonstrates neurotropism, and neurological complications from infection are well described.

The best-known is congenital microcephaly, one of multiple potential neurological complications of congenital Zika infection.

Maternal-to-fetal transmission during maternal Zika infection is likely very frequent, but the reported risk of subsequent congenital abnormality is variable, ranging from 6% to 40% for any abnormality and 1% to 5% for microcephaly.

Birth defects are exclusively associated with the Asian genotype; it is hypothesised that the more virulent African strain may instead lead to fetal loss.

In adults, Zika has also been associated with Guillain-Barré syndrome, meningitis, encephalitis, and transverse myelitis.

848
Q

A man presents after a fishing trip to Zimbabwe. He reports a motile, large firm mass, initially in his neck. It goes away then 3 weeks later presents in his groin. It is itchy and serpinginous. Bloods show eosinophilia. Otherwise nil o/e. What is the likely diagnosis and ddx?

A

Gnathosomiasis caused by Gnathostoma spinigerum

Look for: mobile subcut mass, intermittent creeping eruptions, eosinophilia (may have N&D&V, fever, malaise for 2-3 weeks before cutaneous or visceral manifestations)

Acquired by eating raw fish and other food items -> most common in SE Asia and Latin America
Nematode, zoonotic, foodborne

Tx: Albendazole 21 days /Ivermectin 2 days

Big risk: migration of nematode to brain or spinal cord -> eosinophilic meningitis, SAH, cranial neuritis, painful radiculomyelitis

Ddx:
- Strongyloides stercoralis
- Sparganosis
- Rarer: ectopic fascioliasis, Migratory myiasis

849
Q

Life cycle of gnathostoma

A

adult worms infect GI tract of felines and canines

eggs excreted in faeces get into water

first stage larvae hatch -> infect small freshwater crustaceans

then other animals become intermediate hosts via heating raw or undercooked meat of intermediate hosts eg freshwater fish, crustaceans, crabs, shrimp, frogs etc

850
Q

Diagnostic investigations for schistosomiasis?

A

Microscopy for Eggs
Stool or Urine
Confirms active infection
Limited sensitivity; needs egg excretion, ie can NOT always see Ova in chronic infection

Serology (Antibody)
Blood
Early detection, non-endemic cases
Cannot distinguish past vs. active

Antigen Detection
Urine or Blood
Indicates current infection
Lower sensitivity for some species

PCR
Blood, Urine, Stool
High sensitivity, species-specific
Requires specialized lab

Tissue Biopsy
Tissue (bladder, rectum)
Confirms diagnosis in chronic cases
Invasive, used selectively

Imaging
Bladder, Liver, Spleen
Assesses organ damage in chronic cases
Not diagnostic on its own
USS in chronic cases can be investigation of choice -> eg in urogenital cases can demonstrate hydronephrosis and bladder wall abnormalities
Also cystoscopy shows sandy patches and uraemia

851
Q

What type of inflammation is caused by Schistosoma eggs?

A

Granulomatous

852
Q

What type of Schistosoma is found in Lake Malawi?

A

Schistosoma haematobium -> causes urogenital schistosomiasis

853
Q

Clinical manifestations of infection with schistosoma haematobium?

A

Granulomatous inflammation of bladder wall and ureteral mucosa
Obstructive uropathy
Hydronephrosis
Recurrent bacterial pyelonephritis
End stage renal disease
SCC bladder

854
Q

Examples of Rickettsial diseases?

A

Ehrlichia

Anaplasma

Coxiella burnetii = Q fever

Rickettsia rickettsii = Rocky Mountain spotted fever

855
Q

Ddx for localised swelling with scab in middle?

A

Eschar - rickettsial disease, expect systemic symptoms

Cutaneous anthrax

Insect bite

Folliculitis

Myiasis -> infection with fly maggots (slow growth of boil, no lymphadenopathy, no systemic symptoms, localised itch)

856
Q

What is myiasis?

A

Infestation of live humans and vertebrate animals with larvae (maggots) of flies -> feed on host’s dead or living tissue, liquid body substance or ingested food

Cutaneous -> common manifestation -> red lesion with scab in middle, systemically well

Two types:
- Africa = caused by larvae of Cordylobia anthropophaga (Tumbu fly or Mango fly)
—-> Adult female flies deposit eggs on sandy ground or on damp laundry spread etc -> humans wear contaminated clothes without hot ironing or lie on ground -> larvae hatch and burrow into skin -> over 2/3 weeks developing larvae cause itch and ‘blind boil’
NB: lesions are usually sterile as larva oozes with antibacterial properties

Tx: petroleum over wound -> starves larva of air and eventually comes to surface of skin -> pull it out. Self-limiting regardless as mature larva has to leave host and pupate elsewhere.

  • Central/South America = Dermatobia hominis (human botfly)
    —-> Eggs laid directly on exposed skin + ova also deposited on blood-sucking insects eg mosquitoes, flies, ticks -> conveyed to human host
857
Q

A 50 year old male patient is brought to your hospital in South Africa with a swollen right leg. He says that he was bitten by a snake four hours previously whilst walking barefoot, after dark, along a path near his home. There are clear puncture marks on his right foot and his right foot is oedematous to just below the knee.
The snake most likely to have bitten him is:

A

Puff adder (Bitis arietans)

858
Q

A man is admitted from an informal settlement on the edge of a large city. He is thin, dirty and smells of alcohol.
On examination he is ill and emaciated, his pulse is 98 beats per minute and regular, and his BP is 90/50 mm Hg. He has central cyanosis, widespread basal lung crepitations, ankle oedema and 3 cm soft hepatomegaly. Which of these is the most important drug to give?

A

Thiamine

Wet beri beri

859
Q

What is wet beri beri? How does it present?

A

Thiamine deficiency

Causes:
Inadequate dietary intake, often in populations consuming polished rice.
Increased thiamine demand (e.g., pregnancy, lactation).
Malabsorption issues (e.g., gastrointestinal disorders, alcohol dependence).

Symptoms:
Cardiovascular:
Edema (swelling in legs and feet).
Rapid heart rate (tachycardia).
Low blood pressure (hypotension).
Possible heart failure.
Neurological: Some patients may experience peripheral neuropathy.

Diagnosis:
Based on clinical symptoms and dietary history.
Laboratory tests can measure thiamine levels but are not always necessary.

Treatment:
Thiamine Supplementation: Immediate oral or intravenous administration of thiamine.
Dietary Changes: Encourage consumption of thiamine-rich foods (whole grains, legumes, nuts).

Prevention:
A balanced diet rich in thiamine.
Education about thiamine’s importance, especially in at-risk populations.

860
Q

Principles of a ventilated improved pit latrine

A

The slope of the roof should trap the prevailing wind to allow air to circulate through the latrine

The pipe is designed to trap flies and other insects which are attracted to the light and then trapped by the fly screen which you can see at the top of the pipe, as well as reduce smells if the airflow is adequate.

Most sources would say that 15m or 50 feet is the lower limit of distance from a water source.

The sides of the pit are normally sealed to prevent collapse/undermining by burrowing animals. The bottom is left unsealed so that liquid can seep away- sealing it would cause the latrine to fill up with liquid and flood.

Constructing a latrine on a slight mound is preferred so it does not flood easily with rain or groundwater

861
Q

Appearance of meningococci on blood film?

A

intracellular Gram negative cocci in pairs (diplococci)

862
Q

Is B.bacilliformis or Carrions disease inside or outside cells?

A

Bartonella is intracellular

863
Q

Gram-negative bacilli loose in the blood film which have a bipolar(safety pin) appearance?

A

Yersinia pestis

864
Q

What type of diet is pellagra a feature of?

A

Maize

865
Q

Perifollicular haemorrhages are a feature of what disease?

A

scurvy - check for loss of teeth -> feature of the weakening of connective tissue

866
Q

Causes for reduction in maternal mortality since 1940s?

A

Handwashing
RCOG
Antiseptics
Antibiotics
RCM
NHS
oxytocin
Training
Contraception
MgSO4
Abortion law
Living standards
Safer anaesthesia

867
Q

What are the definitions of maternal deaths?

A

Direct: As a consequence of a disorder specific to pregnancy e.g. Haemorrhage, pre-eclampsia, genital tract sepsis

Indirect: Deaths resulting from previous existing disease, or diseases that developed during pregnancy, and which were not due to direct obstetric causes but aggravated by pregnancy e.g. Cardiac disease, other infections (sepsis)

Coincidental: Incidental/accidental deaths not due to pregnancy or aggravated by pregnancy e.g. Road traffic accident

Late: Deaths occurring more than 42 days but less than one year after the end of pregnancy

868
Q

Worldwide 75% of maternal deaths are due to

A
  • Severe bleeding (PPH)
  • Infections (usually after childbirth)
  • High blood pressure during pregnancy
  • Delivery complications
  • Unsafe abortion
869
Q

Reasons for insufficiency of care wrt maternal mortality?

A
  1. Health system failures
    ○ Poor quality of care
    ○ Insufficient numbers and training of healthcare staff
    ○ Lack of essential resources/medial supplies
    ○ Poor accountability
  2. Social determinants
    ○ Income
    ○ Nutrition
    ○ Education
    ○ Race and ethnicity
  3. Harmful gender norms
    ○ Structural and individual
  4. External factors
    Instability of health systems eg conflict, climate, humanitarian crises
870
Q

What is the three delays model with respect to care?

A

1) Delay in decision to seek care

2) Delay in reaching appropriate care

3) Delay in receiving adequate care

871
Q

Global strategies to address maternal morbidity and mortality:

A
  • SDGs (2015-2030) - under no.3, target 3.1 to reduce MMR <70 maternal deaths per 100,000 livebirths by 2030

Strategies toward ending preventable maternal mortality WHO 2015 (5 objectives, progress updated 2021)

872
Q

WHO’s five strategic objectives for ending preventable maternal mortality?

A

Address inequities in access to maternal health services.

Ensure universal health coverage for comprehensive maternal and reproductive health.

Address all causes of maternal mortality and related disabilities.

Strengthen health systems to meet the needs of women and girls.

Improve measurement, data quality, and use of maternal health data.

873
Q

WHO recommendations for antenatal care 2017? WHO advised schedule?

A

Recommended interventions in five
categories
What?
* Antenatal nutrition
* Maternal and fetal assessment
* Preventive measures
* Management of common physiologic symptoms in pregnancy

How?
* Health system-level interventions to improve the utilization and quality of ANC

Eight contacts
* One in first trimester
* Two in second trimester
* Five in third trimester

  • Tailored to individual country context
  • What is included at each contact
  • Who it is provided by
  • Where it is provided
  • How it is provided and co-ordinated
874
Q

WHO recommendations for antenatal care wrt
- Routine antenatal nutrition
- Maternal and fetal assessment
- Preventative measures
- Common physiological symptoms of
pregnancy
- Health system interventions
- Community based interventions

A
  • Routine antenatal nutrition
  • Promote healthy eating and physical activity
  • Iron and folic acid supplements
  • Calcium supplements for populations with low dietary intake
  • Vitamin A supplements or populations with high deficiency rates
  • Maternal and fetal assessment
  • Check haemoglobin and urine culture using available methods
  • Ask about intimate partner violence if provider is trained to ask/respond
  • Routinely screen all women: for gestational diabetes, tobacco and substance misuse, HIV and syphilis
  • Screen for TB in high prevalence areas
  • Assess fetal growth by SFH or abdominal palpation
  • Provide ultrasound before 24 weeks
    gestation
  • Preventative measures
  • Treat asymptomatic bacteriuria
  • Provide anti-helminthic treatment in endemic areas
  • Vaccinate against tetanus
  • Provide intermittent preventative treatment and insecticide treated nets in malaria endemic areas
  • Common physiological symptoms of
    pregnancy
  • Offer pharmacological
    and non-pharmacological treatments for common pregnancy symptoms
  • Health system interventions
  • Woman-held case notes improve quality of care and pregnancy experience
  • Midwifery-led continuity of care in settings with robust midwifery programmes
  • Task-shifting of ANC activities
  • Support to recruit and retain health workers in rural/remote areas
  • Community based interventions
  • Improve communication and support for pregnant women
  • Increase birth preparedness and complication readiness
  • Increase use of antenatal services and
    skilled care at birth
  • Increase male involvement
  • Build partnership with traditional birth attendants
875
Q

HIV and Malaria contribute to how many maternal deaths globally?

A

25%

876
Q

WHO recommendations for management of threatened preterm labour?

A
  1. Tocolysis
    - 24 – 33+6/40
    - CI: PV bleeding, placental abruption, intrauterine infection
    - To allow antenatal corticosteroids OR transfer to a
    facility for care of preterm infant
    - Nifedipine 20mg, followed by 10mg 6 hourly
  2. Antenatal corticosteroids
    - 24-33+6/40
    - If high chance of labour within 7 days (PPROM, cervix ≥3cm dilated or ≥75% effaced)
    - Or planned preterm birth
    - IM dexamethasone: 6mg, 12 hourly, 4 doses.
    - IM betamethasone 12mg, 24 hourly, 2 doses.
    - Maximum benefit after 24 hours to 7 days.
  3. Antibiotics for PPROM
    - Any gestation <37/40
    - Erythromycin 250mg, qds, 10 days or until birth
    - Reduce risk of chorioamnionitis and NEC
  4. Magnesium sulphate
    - 30% reduction in cerebral palsy <32 weeks
    - 4g loading and 1g/hour maintenance
    - If delivery anticipated within 24 hours
  5. Delayed cord clamping
  6. Thermal care
    Plastic bag/wraps for stabilisation and transfer
877
Q

Preterm birth:
- Definitions
- Prevalence
- Causes
- Prevention

A

Preterm birth
* extremely preterm (<28 weeks)
* very preterm (28 to 32 weeks)
* moderate to late preterm (32 to 37
weeks)

  • 5-18% of births globally
  • Increased burden in southern Asia and sub-Saharan Africa
  • 13.4 million babies in 2020
    Leading global cause of perinatal mortality (30%)

Causes:
* Idiopathic (cause unknown)
* Iatrogenic (pregnancy complications
eg. PET, growth restriction)
* Preterm prelabour rupture of
membranes (PPROM)
* Infection (intra- and extra-uterine)
* Bleeding (APH)
* Multiple pregnancy

Prevention:
* Optimal antenatal care
* Nutrition and lifestyle
* Identify and treat anaemia, asymptomatic bacteriuria
* USS to determine GA and multiple pregnancy
* Cervical length monitoring
* Progesterone
* Cervical cerclage
* (Rescue cerclage)

878
Q

Anaemia in pregnancy
- Definition
- Prevalence
- Causes
- Complications (maternal and fetal)
- Management

A

Definition:
Hb <110g/l in first trimester
<105g/l in second and third
trimesters
<100g/l postpartum

Prevalence:
Around 40% globally
* Highest in SE Asia and Africa
Second leading cause of disability globally

Causes:
* Pregnancy physiology
* Nutritional deficiency (90% of cases)
* Parasitic infection
* Other infections (TB, HIV, malaria)
* Chronic diseases
* Acute/chronic blood loss
* Haemoglobinopathies

Complications:
Maternal:
* Symptoms (fatigue, irritability,
generalized weakness, shortness
of breath)
* Reduction in immune function
(increased susceptibility and
severity of infection)
* Placental abruption
* Post partum haemorrhage
* Increased maternal mortality

Fetal:
- Preterm birth
- LBW
- Increase in infant mortality

Management:
Oral iron supplementation
* Ferrous sulphate 200mg od
* Ferrous fumarate 210mg od
* Expect 1g/week Hb increase

Parenteral iron
* If not tolerating oral or rapid
correction of Hb needed
* Anaphylaxis, skin staining

Blood transfusion
* Hb <6g/dL (<8g/dL if PPH risk)

Folic acid
* 5mg od

Vit B12
* IM or oral cyanocobalamin
* 250-1000mcg alternate days, 2
weeks

879
Q

Hypertensive disorders of pregnancy
- Definition
- Prevalence
- Causes
- Complications (maternal and fetal)
- Management

A

Pre-existing HTN
- Essential
- Secondary to renal disease
- Diabetes
- Systemic disease
- Cardiac causes

Pregnancy induced HTN

Pre-eclampsia
- Pregnancy specific: HTN + proteinuria
- Most common in first pregnancy

Pre-eclampsia, eclampsia, HELLP
* Any organ system may be affected
* Most commonly presents - headache, hypertension, proteinuria
* Fitting in pregnancy should be managed as eclampsia until other
proven cause

Recurrence rate of 10-40%

Complications
- Super-imposed pre-eclampsia
- Preterm birth
- Fetal growth restriction
- Placental abruption
- Perinatal death
- Maternal stroke

Management:
* Prompt and careful control of hypertension
* Meticulous fluid balance with avoidance of overload
* Seizure prophylaxis with magnesium sulphate
* Watch platelets
* Renal function
* May develop coagulopathy
* Planned delivery on the best day is the best way
* Stabilisation before any transfer
* Postpartum vigilence

880
Q

Prevention of HTN in pregnancy? Who is at higher risk?

A
  • Check BP and urine at every point of contact in pregnancy to screen for pre-eclampsia and manage any pre-existing HTN
    Reduce risk by low dose aspirin 150mg from 12-36 weeks for women considered high risk

HIGH
Chronic Hypertension: Pre-existing high blood pressure before pregnancy.
History of Preeclampsia: Previous pregnancies complicated by preeclampsia.
Autoimmune Disorders: Conditions such as lupus or antiphospholipid syndrome.
Kidney Disease: Chronic kidney issues raise the risk of hypertensive complications.
Diabetes: Both Type 1 and Type 2 diabetes are risk factors.
Multiple Pregnancy: Carrying twins or more increases the risk.

LOW
First Pregnancy: Higher likelihood in first-time mothers.
Age: Being under 20 or over 35.
Obesity: Higher BMI increases risk.
Family History: Family members with preeclampsia or hypertension.
Personal History of Conditions: Previous gestational hypertension or related issues in past pregnancies.

881
Q

Key obstetric emergencies that affect mother (features and management):
Maternal collapse
Haemorrhage = APH/PPH
Sepsis

A

Maternal collapse
- Consider uterine manual displacement and peri-mortem C-section

Causes:
4H’s and 4T’s, particularly
- Hypovolaemia
- Hypoxia
- VTE
- Toxicity eg LA, MgSO4
Other:
- Eclampsia
- Pre-eclampsia
- Intracranial haemorrhage
- Amniotic fluid embolus
- High spinal anaesthesia
- Peripartum cardiomyopathy
- Aortic disection

APH
RFs:
Placental praevia
Placental abruption
Uterine rupture

Mx:
ABC
Bloods: FBC, X match, Coag, Kleihauer
Deliver fetus
Prepare for PPH

PPH
‘Empty uterus does NOT bleed’

RFs:
* Retained placenta (tissue)
* Adherent membranes or placental fragments (tissue)
* Uterine rupture (tone and trauma)
* Uterine inversion (tone)
* Atonic uterus (tone)
* Genital tract trauma – cervix or vagina (trauma)
* Coagulopathy – PET, abruption, AFE, sepsis (thrombin)

Mx:
* ABC
* Call for help
* State nature of emergency
* Depends on clinical situation
* Uterotonic medications
* May need urgent transfer to theatre for manual removal of placenta
* Urgent suturing
* Intrauterine pressure with balloon
* Laparotomy and compression of uterus
* Decision for hysterectomy saves lives

In general for bleeding at time of C-section:
* A contracted empty uterus does not bleed
* Empty the uterus
* Step-wise approach to atony
* Give uterotonic drugs
* Pressure, pack, platelets, prayer, patience
* Consider B Lynch suture
* Intrauterine balloon
* Hysterectomy

SEPSIS
- sepsis 6
Women should not feel flu like after birth + young fit women decompensate late
?GrpA strep

Mx
* Avoid NSAIDs
* IV antibiotics as soon as possible
* Full thorough examination ?sepsis source

882
Q

Key obstetric emergencies that affect baby (management for each):
* Breech delivery
* Shoulder dystocia
* Cord prolapse

A

Breech mx:
* Delay pushing until breech is distending the introitus
* Lithotomy
* Episiotomy
* Hands off approach
* Do not squeeze fetal abdomen
* Pelvic grasp and rotate to aid delivery of arms
* When nape of neck appears allow baby to hang down
* MSV to flex and deliver head
* Forceps if head difficult to deliver – assistant to hold baby UP

SD:
Recognise early - ?turtle sign/failure to restitute
* Call for help
* State the nature of the emergency
* Discourage pushing
* McRoberts manoeuvre to maximise size of pelvic outlet
* Routine axial traction
* Suprapubic pressure to try to dislodge anterior shoulder
* Internal rotational manoeuvres or delivery of posterior arm
* Ready for neonatal resuscitation
* Prepare for PPH
* Debrief the mother

Cord prolapse:
ELEVATE THE PRESENTING PART

RFs:
- Malpresentation/unstable lie
- Multiparity
- Polyhydramnios
- Prematurity

Mx:
* State the nature of the emergency
* Is the fetus viable?
* Is the cervix fully dilated?
* Turn off any oxytocin and consider tocolysis
* Manually elevate presenting part
* Catheter with 500ml into bladder
* All fours
* Monitor fetus and transfer for delivery by Caesarean
* Release bladder prior to skin incision
* Debrief the patient

883
Q

Management of uterine atony?

A
  • Bimanual compression
  • Give uterotonics
  • Ensure bladder is empty – indwelling catheter
  • Consider EUA and intrauterine balloon
  • Consider laparotomy
  • Slow 5u IV oxytocin
  • Ergometrine 0.5mg IM or slow IV (not if hypertension)
  • Oxytocin infusion
  • Carboprost 0.25mg IM every 15 mins to maximum 8 doses
  • 1mg misoprostol (5 tablets)
  • Tranexamic acid (in context of active bleeding)
884
Q

Features of (what, relevant RFs, mx)
- Placental praevia
- Placental abruption
- Uterine rupture

A

Placental praevia
Placenta implanted totally or partially in lower segment of uterus
- Recurrent bright red bleeding without pain
- Fetus not usually compromised unless major blood loss
Often high presenting part or malpresentation
RFs:
More common in multiple pregnancy and C-section

Mx:
Do NOT do a vaginal examination
Think about C-section
Entry and haemorrhage control

Placental abruption
Separation from the placental bed of a normally sited placenta
Tender uterus/fetal distress
- Unknown cause
- Usually presents with pain
- Bleeding may or may not be revealed

Complications:
- Fetal compromise
- Maternal shock and DIC

Mx:
Vaginal delivery not necessarily CI

Uterine rupture
Occurs in labour
Presents with pain, bleeding, fetal distress, maternal collapse

RFs:
RFs:
- VBAC
- Multiparity
- Obstructed labour
- Trauma

Mx:
ABC
Theatre
GA if possible
C-section with possible hysterectomy

885
Q

Gynaecological emergencies:

A

Pregnancy related:
o Maternal collapse
o Septic abortion/TOP
o Miscarriage
· Threatened/missed
§ Closed cervix
· Incomplete
§ Open cervix, Ectopic pregnancy

· Heavy menstrual bleeding
· Cervical cancer
§ Rarely present as emergencies but needs to be considered
· Trauma: Assault, secondary to unsafe termination, Non-obstetric/gynaecological

886
Q

Management of gynae emergencies:
- Ectopic pregnancy
- Sexual assault
- Miscarriage (threatened, missed, ongoing, septic)
- Heavy menstrual bleeding
- Sepsis after unsafe abortion
- PID

A

· Ectopic pregnancy
○ Salpingectomy -> Usually mini-laparotomy in low resource settings

· Sexual assault
○ Assess injuries including non-gynaecological
□ Documentation and sampling needs to occur before repair/theatre
○ Risk of pregnancy
○ Emergency contraception
○ STI risk
□ HIV PEP
□ HBV vaccination
□ Consider Abx if no follow-up available
○ Mental health assessment
□ Consider admission if needed

· Miscarriage
○ Threatened
□ Reduce activity –> either threat passes or abortion inevitable
□ Consider infectious cause (e.g. malaria, STI) –> treat
□ Analgesia
○ Missed
□ Uterine evacuation not urgent if no signs of infection, no heavy bleeding
□ Uterine evacuation
◊ Misoprostol 600mcg SL or 800mcg PV
◊ Manual vacuum aspiration (MVA) under LA
○ Ongoing/incomplete
□ Analgesia
□ Remove POC from vagina and cervix
◊ If RPOC may need misoprostol/MVA
□ Consider underlying cause
□ Iron/folate supplementation
○ Septic
□ As above
□ Remove foreign bodies
□ Expedite uterine evacuation
□ Abx

· Heavy menstrual bleeding
○ NSAIDs
○ TXA
○ COCP
○ Oral progesterone only
○ Gynaecology specialist referral –> IUD

· Sepsis after unsafe abortion
○ Often identified late, fail treatment
○ Examine for foreign body
○ Broad spectrum Abx

· PID
○ Indications for admission
□ High fever >38
□ HR >110
□ Rebound tenderness
□ Palpable pelvic mass
□ Non-responsive to treatment or unable to take PO medications
○ US for tubo-ovarian abscess
○ Broad spectrum Abx
○ Contact tracing and treatment
○ HIV testing
○ Advise re: safe sexual practices

887
Q

Genitourinary fistulae
- Definition
- Pathophysiology
- Risk factors

A

An abnormal opening between the vagina and other nearby organs in the pelvis

2million women globally but decreasing incidence

Iatrogenic fistulae are on the rise (ie used to be more ischaemic as described below), occur due to:
§ Hysterectomy, caesarean hysterectomy, caesarean delivery
§ Gaps in healthcare worker density
§ Healthcare worker skillset/training
§ Infrastructure

Pathophysiology
o Obstetric fistula
· Used to be major cause
· Usually occurs in second stage
· More common in primiparous women
· Fetal head compressed between pubic symphysis, sacrum –> oedema of (anterior) vaginal wall –> extreme pressure necrosis
§ Causes widespread ischaemia of surrounding tissues –> sloughing –> fistula formation
§ Can occur even if LUSCS performed
§ Often takes several days after delivery for diagnosis –> may not be diagnosed before discharge

· Risk factors
§ Prolonged or obstructed labour
§ Late referrals from community settings
§ Distended bladder
§ Use of oxytocin in second stage
§ Failed instrumental
§ Destructive delivery after prolonged labour
§ Uterine rupture/pre-rupture (Bandl’s Ring)

Teenagers eg as in child marriage, are at risk
Low education, rural location

888
Q

Genitourinary fistulae
- Complications

A

Complications
· Urinary/faecal leakage
· Abnormal vaginal discharge
· Tissue damage/irritation
· Pyelonephritis
· Infertility

§ Nerve damage –> foot drop
§ Perineal injury
§ Urethral loss/stress incontinence
· Hydronephrosis, renal failure
§ Rectal atresia, anal sphincter incompetence
§ Cervical destruction
§ Asherman’s syndrome –> amenorrhoea
§ PID
§ Secondary infertility
§ Vaginal stenosis
§ Osteitis pubis
§ Sociocultural consequences

889
Q

What is the single leading cause of perinatal mortality?

A

Preterm birth

890
Q

How does the epi of genitourinary fistulae vary according to economic setting?

A

· High income countries
§ Gynaecological surgery
· Usually multiple operations
§ Pelvic malignancy
§ Complications of RTx

· Middle income countries
§ Iatrogenic
§ Contribution from cervical cancer, obstructed labour, genitourinary TB

· Lower income countries
§ Relatively high rates of ongoing obstetric fistula but improving
· Improving maternal mortality improves morbidity
Increasing rates of iatrogenic fistula

891
Q

Prevention tools for obstetric fistulae vs iatrogenic

A

Obstetric fistula
· Primary prevention
§ Health promotion
§ Planned pregnancies
· Birth spacing
· Contraception, abortion care
· Prevention of child marriage
· Addressing violence
Community awareness

· Secondary prevention
§ Antenatal care
§ Skilled personnel at birth
§ Use of partograph
§ Identification of obstructed labour
§ Immediate referral
§ Bladder care during labour
· Consider catheterisation if needed
· Can consider catheterisation post-partum after obstructed labour

· Tertiary prevention
§ Access to emergency obstetric and neonatal care including safe emLUSCS

Iatrogenic fistula:
· Requires prevention of surgical error
§ Reducing non-indicated C-sections
§ Restricting surgery to authorised, registered and trained surgeons

892
Q

How do you diagnose and treat genitourinary fistulae?

A

DIAGNOSIS
Methylene blue: via IDC into bladder
Vagina packed with gauze, observe ?dye leakage
· Only diagnoses vesicovaginal/urethrovaginal fistula

Imaging

TREATMENT
o Leave IDC (silicone) in for 4/52
· Can be observed in community
· Weekly review for hydration status, signs of infection
· Repeat methylene blue test after 4/52
§ If neg -> remove catheter, consider bladder training
§ If pos -> remove catheter and refer
· Don’t leave IDC in for >6/52 total

o Referral to specialist centre for all vesico/urethrovaginal fistulae
· Take details including contact details (including of alternative contact)

o Basic hygiene and care advice
· Drink 4L/day of water
o Offer contraception
o If large rectovaginal fistula with severe contamination, irrigation with diluted povidone/betadine after every stool
· Treat diarrhoea

o If small rectovaginal fistula, may close spontaneously
· Sitting baths, regular intimate toileting may prevent infection

893
Q

Characteristics of cholera outbreak in terms of settings?

A

· Rural settings
§ High population number, low density
· Low attack rate (0.1-2%)
· Slow peak (1-3m)
§ Lasts 3-6m
§ Fatality rate <5%

· Urban settings/slums
§ High population number, high density
· High attack rate (1-5%)
§ Peak 2-8 weeks
§ Duration 2-4m
§ Fatality rate 2-5%

· Closed settings (refugee camps)
§ Low population number, high density
· High attack rate (1-5%)
· Rapid peak (2-4w), short duration (1-3m)
- Fatality rate <2%

894
Q

Cholera
- Cause
- Epi and transmission
- Pathophysiology
- Symptoms
- Diagnosis
- Complications

A

Cause

Vibrio cholerae bacteria (serogroups O1 and O139 most common for epidemics)
Highly motile, rapidly reproducing

Epidemiology

Endemic in parts of Africa, South Asia, and Southeast Asia; outbreaks occur after natural disasters or in settings with poor water and sanitation infrastructure
Incidence increasing (conflict, climate change (low water stock -> more conc organisms), vaccine shortage)

Transmission

Primary: Fecal-oral route -> ingested via infected water source
Requires large ingestion load to cause illness
Secondary:
Contaminated water or food
Flies
Soil
Hand hygiene -> food contamination
NB - Can have asymptomatic carriers

Pathophysiology

Ingested –> multiply in stomach –> toxin secretion
Massive secretion of water and electrolytes in the intestines

Symptoms

Incubation: can be convalescent for 14 days
Mild: Often asymptomatic or mild diarrhoea
Severe: Profuse (up to 15L/day), “rice-water” diarrhoea, vomiting, rapid dehydration, shock, acidosis

Diagnosis

Clinical: Based on symptoms in endemic areas
Laboratory: Stool culture, rapid antigen tests (e.g., dipstick or PCR for V. cholerae detection)

Complications

Severe dehydration, electrolyte imbalances, shock, acute kidney injury, death if untreated
Case fatality highest in pregnancy, in general up to 50% without treatment, <1% with treatment

895
Q

Management of Cholera cases and outbreaks

A

Rehydration: Primary treatment, ORS or IVF (if severe)
Antibiotics: in severe cases to shorten illness duration, consider Doxy, resistance increasing
Hygiene: Clean water, sanitation, handwashing
Vaccination: Oral cholera vaccines (e.g., Dukoral, Shanchol) available in high-risk areas

Evaluation: need to collect data on CFR, weekly incidence rate, attack rate

Outbreak management:

Case management

Isolation and treatment
· Decentralisation to keep people in affected communities
· Treatment separated from existing healthcare facilities –> remove transmission risk
· Cholera treatment centres: large, autonomous, 24/7, offer IV and PO rehydration
· Cholera treatment units: simpler, smaller, 24/7, offer IV and PO rehydration
· Oral rehydration point: outpatient, in-hours, simple cases, referral pathway to CTU/CTC

Training of healthcare workers

Use of protocols
· Triage and admission: diagnosis, assessment of hydration status, initial treatment
· Hydration: compensating for losses, close supervision (every 30min), zinc supplementation for children <5yo
· Complementary therapy: Abx, nutritional

Early case detection and referral
· Active case finding

WASH in treatment facilities
§ Supply of chlorinated water
§ Adequate sanitation and waste management
§ Strict IPAC measures
§ Management of bodies

896
Q

Methods for the prevention of Cholera

A

o Vaccination (oral)
§ Appropriate >1y, including pregnant women
· Low efficacy in children (43% <5yo)
§ 2 doses, 14d apart
· Immunity 7-10 days after 1st dose
§ Immunity lasts at least 6 months, up to several years (not lifelong)
· Protected for 3-5 years after 2 doses
§ Also reduces transmission in healthy carriers
§ Should be stored at 2-8 degrees but strict cold chain not required
§ Still effective after 3-4 days at 40 degrees
§ Can vaccinate reactively or preventively

o Health promotion
· Consideration to lack of literacy –> need oral delivery

o Hygiene
· Hand & household hygiene
· Food hygiene (eat immediately –> decrease risk of contamination after cooking by flies)
· Safe burial/funeral practices -> avoid touching, disinfection, burial 1.5-2m deep, avoid water sources

o Sanitation
· Disposal of human waste + waste water

o Access to water
· 15-20L/person/day -> needs increase with cholera
· Chlorination (in line, batch, bucket -> must cover, inactivated in sunlight, aquatabs)

o Risk stratification
· Ring strategy/case area targeted intervention
§ Households surrounding cholera cases at increasing risk with increasing proximity to affected household
§ Implement WASH interventions, disinfection, vaccination to reduce inter-household transmission

897
Q

When is greatest risk of newborns for death globally (including NHS)?

A

First week
Esp intra-partum

898
Q

Key causes of under 5 mortality:

A

1 Prematurity
2 LRTI/Pneumonia
3 Intrapartum related eg birth asphyxia
4 Diarrhoea
5 Neontal infection

899
Q

Most important way to prevent neonatal deaths?

A

Family planning

900
Q

Challenges for neonatal care

A
  • Assessing gestational age
  • Providing warmth
  • Feeding and fluids
  • Respiratory support
  • Infection prevention, recognition and treatment
  • Different exposures; context and care settings
  • Care largely supportive; the majority do not have access to NICU
  • Minimal capacity for investigations
  • Low nursing ratios, minimal monitoring
  • Risks of compounding brain injury; cooling not universally implemented
  • Limited access to fit-for-context equipment ie. pumps
  • Supply chain challenges and drug outages
  • Inconsistent electricity & water supplies
901
Q

Epi of neonatal health/deaths

A

Child mortality reducing:
* 12.6 → 5 million from 1990 to 2020
Newborn deaths still lag:
* 2.4 million newborn deaths, ¾ of these in the first week
* 47% of all under-5 mortality
* Preterm birth and birth complications are the leading causes of death under-5

250 million children –are at risk of stunted growth and poverty and unlikely to
fulfil developmental potential
An estimated 53 million children with developmental disability

902
Q

What are neonatal conditions associated with and what do you have to think about longterm? What is a plan with targets for neonatal care and what is the neonatal mortality rate target?

A

· ‘Every Newborn Action Plan’ lays out strategic objectives and ambitious targets to
reduce neonatal mortality NMR or <12 per 1000 births
· Neonatal conditions are associated with an increased risk of developmental delay
and disability
· Important to think ‘Beyond Survival’ … to survive AND THRIVE including early
intervention for young children with developmental disability

903
Q

What is amongst the leading causes of child mortality and responsible for almost half of all deaths under 5 years?

A

Neonatal conditions

904
Q

When is the highest risk of death for a baby? What are some leading causes of neonatal deaths?

A

· The highest risk of death is the day of birth, 75% in the first 7 days (highest risk is in first 24 hrs)
· Prematurity, birth complications and neonatal infections are the leading causes of
neonatal deaths

905
Q

What is a preterm birth?

A
  • Late preterm 34 - <37 weeks
  • Moderate preterm 32 - 34 weeks
  • Very preterm 28 – <32 weeks
  • Extremely preterm <28 weeks
906
Q

Challenges of providing neonatal care in low resource settings?

A

Power, water, space
Assessing gestational age
Providing warmth
Feeding and fluids
Infection prevention, recognition, treatment
Respiratory support
Prevention of apnoea
High patient nurse ratio

907
Q

Definitions of prematurity/baby wrt birthweight? why relevant?

A
  • Low birthweight, LBW < 2500g
  • Very low birthweight, VLBW < 1500g
  • Extremely low birthweight, ELBW < 1000g

A lot of women won’t know LMP etc so actually can’t work out age of babies
This weight method is also an issue as a lot of babies in LMICs are low weight for gestational age

908
Q

Risk factors for preterm mortality

A
  • C-section
  • Born outside hospital
  • Moderate/Severe hypothermia at admission (highest risk factor)
    ○ Hypothermia stops surfactant production, increases respiratory distress
    ○ Use more energy, increasing glucose requirements, can become hypoglycaemic
    ○ Increased risk apnoea
  • Bradycardia at admission
  • Hypoxia at admission
909
Q

Methods in LMICs to prevent hypothermia in preterms/from birth?

A
  • Dry and cover
  • Skin to skin (Kangaroo)
  • Put on hat and nappy
  • Avoid bathing
910
Q

Leading cause of deaths in preterm babies?

A

RDS
Need CPAP

911
Q

Relevance of breastmilk for neonatal care, especially in preterm infants in LMICs? What are some issues with respect to nutrition and postnatal growth failure? Some solutions in low income settings?

A

Compared to formula milk, in preterm infants, breast milk reduces the risk of:
○ Sepsis
○ Necrotising enterocolitis
○ Retinopathy of prematurity
○ Feeding intolerance
○ Neurodevelopmental delay
* Wherever possible choose early & exclusive breast milk diet
How?
Use mum’s milk expressed into cup every 2 hours -> feed by NG to establish feeds, feed by cup and spoon when stable, off CPAP and gaining wait

BUT
- Over 40% of preterms in neonatal units in LMICs have insufficient access to mum’s own milk
- Preterms are at increased risk of postnatal growth failure (PGF)
- PGF increases the risk of neonatal mortality
- PGF also increases risk of subsequent neurodevelopmental delay in surviving preterm infants
- PGF contributes to the high neonatal mortality rate in Uganda

Solutions:
- Nutritional support for mothers
- Educate/encourage acceptability of donor breastmilk
- Lactational support
- Low cost, sustainable milk bank

912
Q

Risk factors for early-onset neonatal sepsis?

A

Maternal Risk Factors
* Fever (>38C) before or during labour
* Foul-smelling or purulent amniotic fluid
* Premature rupture of membranes (>18 h)
* Multiple unclean vaginal examinations
during labor
* Prolonged or difficult labor (>24 h)

Fetal Factors
* low birth weight
* Prematurity
* perinatal asphyxia

913
Q

Case definition for possible severe bacterial infection wrt neonatal infections? What are some sequelae?

A

Case definition
* History of feeding difficulty
* History of convulsions
* Movement only when stimulated
* Respiratory rate > 60 breaths/min
* Severe chest indrawing
* Temperature >37·50C, or <35·50C

Sequelae:
Pregnancy
- Still birth
Neonatal period
- Meningitis
- Sepsis
- Pneumonia
- Tetanus
- Death
Childhood and adulthood
- Impairment
- Neurological and neurodevelopmental issues
- Hearing issues
- Physical and mental health issues

914
Q

Classification of neonatal sepsis and relevant risk factors:

A

Early onset neonatal sepsis
- Occurs within first 72 hours of life (days 0 – 2)
- Often caused by pathogens that the baby is exposed to before or during birth
- Causative organisms may originate from the maternal genital tract
- Unhygienic delivery practices and unclean birth area

Risk factors:
Maternal Risk Factors
* Fever (>38C) before or during labour
* Foul-smelling or purulent amniotic fluid
* Premature rupture of membranes (>18 h)
* Multiple unclean vaginal examinations
during labor
* Prolonged or difficult labor (>24 h)

Fetal Factors
* low birth weight
* Prematurity
* perinatal asphyxia

Late onset neonatal sepsis
- First 29 days

Risk factors:
- NICU admission (ie hospital acquired)
- Invasive medical procedures (IV cannulations; mechanical ventilation)
- Administration of stock solutions during parenteral therapy
- Contaminated equipment, IV or enteral solutions
- Prolonged hospitalisation

For community acquired:
- Poor hygiene practices in home setting
- Inadequate cord care
- Bottle>breast feeding
- Lack of exclusive breastfeeding

915
Q

Key pathogens for early vs late onset sepsis?

A

Early-onset
Group B Streptococcus (GBS)
Escherichia coli
Listeria monocytogenes
Enterococci

Late-onset
Coagulase-negative Staphylococcus
Staphylococcus aureus
Escherichia coli
Klebsiella

916
Q

Prevention tools for
- Maternal infection
- Early onset sepsis/infections
- Late onset sepsis/infections

A

Maternally acquired
* Intrapartum antibiotics
* Penicillin (GBS)
* Azithromycin
* Maternal vaccines
* RSV
* Pneumococcus (RCTs)
* GBS (in development)

Early-onset infections
* Handwashing before delivering and handling the newborn
* Good basic hygiene & cleanliness during delivery
* Avoid unnecessary separation of the newborn from the mother
* Appropriate umbilical care
* Appropriate eye care

Late-onset infections
* Exclusive breastfeeding
* Strict hand-washing (alcohol rub) protocols for all staff and families
* Clean injection practices
* Removing IVF when no longer necessary
* Strict asepsis for all procedures

917
Q

Management strategy for neonatal infections/sepsis?

A
  1. Detection
    • Improved lab support and rapid response -> micro culture = GOLD STANDARD
    • POC diagnostics
  2. Antimicrobials
    • Treatment protocols + IV Abx access
    • Current local/regional epi
    • AM stewardship
  3. AMR
    • Vectors of transmission
    • Surveillance
  4. IPC = infection control measures
    • Environmental and hand hygiene
    • Context specific guidelines
  5. Investment
    • Resources and research
  6. Supportive care
    • NICU admission
    • IVF
    • Temperature control
    • Euglycemia
    • Support for oxygen
      • Blood products
918
Q

Why is AMR an issue for neonatal sepsis?

A

Resistance to WHO-recommended antibiotics (Lancet study:)
* β-lactams 68% (614/904 cases)
* Aminoglycosides (Gentamicin 26% - 83%; Amikacin 0 – 21%)
Resistance to alternative therapeutic options
* Fluoroquinolones (14% – 30%)
* Carbapenems (0 – 4%)
* Piperacillin/tazobactam (7% - 37%)

Wide variation in treatment.
Frequent switching of antibiotics due to high resistance to treatments.

919
Q

Main causes of gram negative AMR bacteria that cause neonatal sepsis in LMICs?

A
  • Klebsiella pneumoniae
  • Serratia marcescens
  • Klebsiella michiganensis
  • Escherichia coli
  • Enterobacter cloacae complex
920
Q

What did the BARNARDs study show with respect to neonatal sepsis?

A

BARNARDs
Showed 4 commonly prescribed Abx combinations for neonatal sepsis:
* Ampicillin–gentamicin,
* Ceftazidime–amikacin,
* Piperacillin–tazobactam–amikacin
* Amoxicillin Clavulanate–amikacin

921
Q

Key African causes of neonatal meningitis?

A

Grp B strep
Strep pneumoniae
Staph aureus

922
Q

WHO treatment of neonatal sepsis?

A

1st line: Gentamicin + Penicillin/Ampicillin
2nd line: Cephalosporin eg ceftriaxone or cefotaxime

923
Q

What did the ANISA study show?

A

Large observational study on neonatal sepsis in Bangladesh/India/South Asia

Key causative organisms were:
* RSV - 5.39
* Ureaplasma - 2.38
* Klebsiella pneumoniae - 1.49
* Escherichia coli - 1.43

924
Q

A indigenous grandmother in America who lives in a crowded family home falls ill. As the community GP you go to see her and she has a widespread crusted skin rash all over her body with dried skin flakes on the bed. She is also NEWs 7 and you are worried about sepsis. What is the likely diagnosis? What are some differentials?

A

Crusted scabies - sacroptes scabiei
Can get superimposed skin infection eg staph aureus or strep pyogenes, also gram negative eg pseudomonas aeruginosa

Ddx:
Skin based eg psoriasis, tinea corporis, skin malignancies eg T cell lymphoma, Sezary syndrome
Nutrition deficiencies eg pellagra, pemphigus, lepromatous leprosy, secondary syphilis

Treat with:
- IV Abx if superimposed bacterial infection
- Ivermectin / benzyl benzoate / keratolytic cream
- Treat family with topical permethrin

925
Q

Gold standard investigation for Pott’s disease?
Some disease features?

A

CT guided percutaneous vertebral or paravertebral aspiration

In endemic settings usually: hx + epi + X-rays *if available

On Spinal radiograph:
- Focal areas of erosions
- Osseous destruction in anterior corners of vertebral bodies
- Involvement of adjacent disc or vertebral body
- Wedging
- Gibbus deformity
- Paraspinal abscess formation

926
Q

Ddx for spinal cord disease in tropical settings

A

Compressive:
- Potts disease (subacute, +/-bladder)
- Metastases (acute/subacute +/-bladder)
- Degenerative spinal disease eg slipped disc

Non-compressive:
- Transverse myelitis (acute, often non-progressive, bladder involvement common)
- Schistosomiasis (acute/subacute, often flaccid paresis, bladder involvement common)
- HIV associated vacuolar myelopathy

927
Q

What are the three most common causes of genital ulceration and relevant features?

A

Herpes simplex
- usually painful
- can have multiple lesions
- usually self-healing (after a few days) in immunocompetent but may be more severe/persist in immunocompromised -> consider tx with aciclovir

Syphilis
- usually single lesion and painless = chancre
- treat with IM BenPen
- serology can be negative in early infection

Chancroid
- Haemophilus ducreyi
- multiple, painful lesions
- common in sex workers/people who go to them
- gram stain not sens/spec -> need to culture at 33 degrees from swabs
- inherently resistant to penicillins, also lots resistant to tetracyclines and sulphonamides
- Tx with azithromycin OR erythromycin OR Ciprofloxacin

928
Q

What enteric pathogen is more common in HIV positive individuals than typhoid fever?

A

Non-typhoidal salmonellae (iNTS)

HIV seems to provide some protection against typhoid fever (weirdly)

929
Q

How does typhoid fever present?
Where is it found?

A

Epi: v common S/SE Asia, medium incidence Latin American and Africa

CANNOT DISTINGUISH Salmonella typhi and paratyphi clinically

Typhoid fever is exclusively human disease -> no animal reservoir. Advise people to wash hands, clean food etc

Incubation Typhoid fever: 10-20 days (range 3-56)
Paratyphoid: 1-10 days

Clinically:
Non-specific eg fever, frontal headache, abdominal pain, constipation (rarely foul-smelling diarrhoea), dry cough
At worse: confusion, myocarditis, toxaemia, intestinal perforation, haemorrhage

Labs: leukopenia + (normocytic anaemia) + thrombocytopenia + slightly elevated transaminases

930
Q

Diagnoses/management of typhoid fever

A

Isolation from stool or urine indicates carrier state -> does NOT prove disease

Ideally need bacteria isolated from BCs or BM

Serological lacks specificity

Start on ABX depending on resistance patterns eg fluoroquinolone like cipro as they have v high intracellular concentrations. If resistance eg in SE Asia then use azithromycin or 3rd gen cephalosporins

If resource constrained setting consider: chloramphenicol, amoxicillin, co-trimoxazole

Treat between 1-2 weeks

931
Q

Vaccines for typhoid fever?

A

Both produce protection of only 70-80%
- parenteral Vi-capsular polysaccharide Ag vaccine
- oral live attenuated vaccine

932
Q

Management of neonatal tetanus

A

Reduce stimulation -> quiet, dark environment

Abx: IV Penicillin +/- metronidazole

Antitoxins - tetanus antitoxin human/equine

Sedative -> chlorpromazine/phenobarbitone

anti-spasmodic -> diazepam etc

Magnesium to reduce spasms

933
Q

Do children who get tetanus need to be vaccinated after the initial episode?

A

Yes = infection does not provide immunity

934
Q

How does localised tetanus present?

A

Onset of weeks
Discomfort around wound
Rigidity
Pain
Weakness
Increased deep tendon reflexes

Can develop into generalised tetanus

935
Q

How does cephalic tetanus present?

A

Incubation period of days
Lower cranial nerve muscles
Facial palsy/stiffness
Trismus
Pharyngeal spasms
Laryngeal spasms, dysphagia, neck stiffness
Paresis of IX/X/III

936
Q

F diagram for sanitation/wash

A

Faeces
(sanitation)
- Fluids
- Fingers
- Flies
- Fields
(safe water)
(hygiene and hand washing)
Food
(hygiene and hand washing)
New host

937
Q

Definition of
- sanitation
- sanitation chain

A

Sanitation: the safe disposal of excreta and human faeces to protect the health and contamination of the environment

Sanitation chain: the safe containment, transport, treatment and disposal/re-use of human excreta

938
Q

What is the sanitation ladder and stages of it with respect to waste?

A

SAFELY MANAGED
Use of improved facilities which are not shared with other households and where excreta
are safely disposed in situ or transported and treated off-site
BASIC
Use of improved facilities which are not shared with other households
eg pit latrines, dry pit latrines
LIMITED
Use of improved facilities shared between two or more households
UNIMPROVED
Use of pit latrines without a slab or platform, hanging latrines or bucket latrines
OPEN DEFECATION
Disposal of human faeces in fields, forests, bushes, open bodies of water, beaches and
other open spaces or with solid waste

939
Q

Epi of lack of access to sanitation

A

Worse in central Africa
- 46% of people do not have safely managed services WW
~20% (1.7 billion) people lack basic sanitation services

940
Q

Stages of faecal sludge management

A

Pit filling
Pit emptying (or not): manually or mechanically
Transport
Waste disposal
Treatment or not
Last two can include: drying beds, anaerobic digestion for biogas production, turn waste into fuel

941
Q

What are the benefits of sanitation?

A
  1. Health benefits
    - Can reduce risk of diarrhoea by up to 25%
    - Reduced risk of: Ascaris, hookworm, Strongyloidiasis, Chlamydia, Trachoma, Schistosomiasis, Nutrition
    - Best is sewered system compared to improved
    • Barreto study 2007: city-wide sanitation intervention reduced childhood diarrhoea by 43% in high risk areas but no effect in low risk areas
      ○ High risk: high baseline prevalence of diarrhoea and lower initial sanitation coverage
      ○ Big investments eg sewer systems are what’s really needed
  2. Privacy, convenience, time, cleanliness and smell
  3. Increases school attendance
  4. Don’t have to withhold food/drinks
  5. Prevent imprisonment by daylight
  6. Protection from harassment and rape
942
Q

What are the stages of the sanitation ladder with respect to water?

A

SAFELY MANAGED
Drinking water from an improved water source that is located on premises, available when needed and free from faecal and priority chemical contamination

BASIC
Drinking water from an improved source, provided collection time is not more than 30 minutes for a round trip, including queuing

LIMITED
Drinking water from an improved source for which collection time exceeds 30 minutes for a round trip, including queuing

U N I M P R O V E D
Drinking water from an unprotected dug well or unprotected spring

SURFACE WATER
Drinking water directly from a river, dam, lake, pond, stream, canal or irrigation canal

Note: Improved sources include: piped water, boreholes or tubewells, protected dug wells, protected springs, and packaged or delivered water.

943
Q

What is the WHO/UNICEF JMP definition for improved water source?

A

A source that by nature of its construction or through active intervention, is protected from outside contamination, in particular from contamination with fecal matter

944
Q

Examples of improved vs unimproved water sources?

A

IMPROVED
Piped water into dwelling In general:
network & tap
Piped water to yard/plot Public tap/ Public standpost Tubewell/Borehole/Hand pump Often with a handpump
Protected Dug well/spring
Protected Spring
Rainwater

UNIMPROVED
(Collected) Surface water
Unprotected dug well
Unprotected spring
Tanker truck/Cart with tank

945
Q

What is the desired maximum protection time for water and why?

A

30mins
More water -> more likely to practice hygiene
If only 10-20L per day -> less likely to use for hygiene eg hand washing, will prioritise it for food, drinking etc

Minimum for “basic” access according to WHO: Improved source & available within 30 minutes (round-trip)

946
Q

Treatment of water methods?

A
  • Chlorination -> residual effect to continue disinfection, does not get rid of Crypto
  • Boiling -> expensive and requires fuel
  • Filtration: slow sand filters
  • UV disinfection (SODIS -> solar) -> Fill up to 2L, corrugated roof to reflect UV back and maintain temp of water

With user methods difficult to know how effective methods are ie were cans left in sun long enough, was enough Cl put in etc

947
Q

The Bradley classification of water-related infections (1972) + interventions

A

Transmission Route, Description, Example
Water-borne
You drink it
Cholera
Improve drinking water quality
Improve sanitation

Water-washed
Person-to-person transmission due to inadequate personal and domestic hygiene
Trachoma, conjunctivitis, louse-
borne typhus
Increase quantity of water
Improved hygiene by increasing access and
availability of water

Water-based
Transmission of infections via an obligatory aquatic host (e.g., snail)
Schistosomiasis, guinea worm
Decrease contact with contaminated water
Control aquatic animals
Reduce excreta entering environment

Water-related insect vector
Transmission by insects which breed in (or bite near) water
Malaria, dengue, yellow fever, African Trypanosomiasis, filariasis
Eliminate breeding sites
Netting, repellent
Keeping people away from breeding sites

948
Q

Definition of hygiene

A

The conditions and practices that help maintain health and prevent spread of disease

949
Q

Handwashing ladder

A

Basic: availability of handwashing facility with soap and water at home

Limited: availability of handwashing facility lacking soap and/or water at home

No facility: no handwashing facility on premises

950
Q

How much water does washing hands use? Does temperature matter? Should you still wash your hands with water alone?

A

Use running water + temperature does not matter
1min with piped water can use up about 5-10L
Most people wash their hands for 10 seconds so this is about 0.5-2L

 Water scarce settings: eco-friendly devices claim to be able to facilitate handwashing with about less than 100ml of water

 Rinsing hands with water and scrubbing thoroughly is better than not handwashing at all and could reduce pathogens by up to 77%

951
Q

What did the GEMs study show?

A

GEMS study: aimed to assess the population-based burden of diarrhoeal disease in children living in sub-Saharan Africa and South Asia

Key pathogens:
- Rotavirus
- Cryptosporidium
- Shigella
- Enterotoxigenic E Coli

952
Q

How does diarrhoeal disease impact health and what are some consequences on long term health?

A
  • Inhibits nutrient absorption → malnutrition, even if sufficient nutrition is consumed
  • Reduces resistance to infections
  • Early childhood development, including stunting and wasting
  • Cognitive development (e.g. language, memory)
  • Lifelong consequences, such as productivity or ability to learn
953
Q

How does WASH affect childhood undernutrition?

A

Inadequate WASH leads to exposure to enteric pathogens
Leading to
- Soil transmitted helminth infections
- Environmental enteric dysfunction (inflammation of small intestine, marked by alternations in gut lining)
- Diarrhoeal diseases

All leading to poor nutritional status
BUT NO EVIDENCE suggesting WASH influences actual nutritional outcomes eg weight for age/height etc

954
Q

What type of bacteria causes tetanus?

A

Clostridium tetani
Anaerobic Gram-positive bacillus
Hardy ubiquitous environmental organisms

955
Q

What does the tetanus toxin do?

A

Disrupts release of inhibitory neurotransmitters, causing
Trismus
Risus sardonicus
Opisthotonos

956
Q

A 7 day old baby comes to a health centre in Ethiopia with a Fever of 38oC

HPc:
Doesn’t handle well
Breath holding episodes
Born in good condition at
home at term 2.6kg
Difficulty opening
Delivered by local old
mouth
woman
Sensitive to light, sound,
Cord cut with a clean blade

What are your differentials?

A

Neonatal tetanus (NNT)
Encephalitis/meningitis
Rabies
TMJ disease
Cerebral malaria
Strychnine poisoning

957
Q

4 challenges to global food security

A

Climate
Conflict
Covid
Chaotic/Corrupt/incompetent leaders, systems

958
Q

Epi of malnutrition

A

4.9 million child under 5 deaths per year -> under nutrition contributed to 45% of these deaths

Global nutrition report 2020
- 1 in 9 people hungry/undernourished -> increase since 2015
- 1 in 3 people overweight/obese

959
Q

Features of malnutrition?

A

Marasmus = wasting:
- Generally wasted
- Thin arms (MUAC)
- Thin face, “old man”
- Ribs visible
- Sunken eyes
- Lack of skin turgor
- (NB assessment hydration!)

Kwashiorkor = oedematous malnutrition:
- Bilaterally pitting oedema
- Apathetic
- Sparse, discoloured hair
- Dermatosis
- Enlarged liver
- Angular chelitis

959
Q

Definition of malnutrition
How to measure it
Classification of malnutrition states (4)

A

Any condition in which deficiency, excess or imbalance of energy, protein or other nutrients adversely affects body function and/or clinical outcome

How to measure:
1) ‘RAW’ measurements (weight, length/height, MUAC)
2) COMBINE to form an INDEX
- Eg height for age, weight for age or BMI
3) COMPARE against a ‘reference’
- Growth curves using age and BMI

Wasting (thinness / marasmus) = acute malnutrition, = low weight-for-height / MUAC
Stunting = chronic malnutrition = low height-for-age
Underweight = acute & chronic malnutrition = low weight-for-age
Micronutrient malnutrition (+/- specific clinical signs)

NB: MUAC only between 6 months to 5 years

960
Q

What is the entitlement approach with respect to malnutrition and what model does it fall under?

A

Socioeconomic reasons for malnutrition. Includes failures such as:
○ Production based eg reduced harvest
○ Trade based (import bottlenecks)
○ Labour based (poverty, unemployment)
○ Transfer based (late delivery of food aid, profiteering)

961
Q

WHO 1999 classification of acute malnutrition

What is the new classification as of 2008 and 2023?

A

Normal range
Weight for height +2 to -2 SDs from median score = Z score

Acutely malnourished includes:
Moderate acute (MAM)
-2 to -3 Z scores
or
MUAC<125mm
(moderate wasting)
No oedema
Can manage supplementary feeds as outpatient

Severe acute (SAM)
<-3 Z scores
or
MUAC<115mm
-> marasmus
Oedema Yes -> kwashiorkor
Tends to need inpatient stabilisation

NB: any bilateral oedema is then classified as severe acute even if weight or height is ok
If a patient slowly deteriorates along the spectrum then their anthropometric status may be worse than their clinical status
If fast deterioration in nutrition then might get matching fast deterioration in clinical status

2008
MAM uncomplicated -> outpatient supplementary feeds
SAM uncomplicated -> outpatient therapeutic feeds
SAM complicated -> inpatient stabilisation

2023
Wasting and oedematous malnutrition

962
Q

How are organ systems affected by malnutrition?

A

Metabolism
- Reduction in metabolic rate and protein synthesis

Heart/CVS
- Output reduced, anaemia (decreased production and increased RBC loss)

Kidneys
- GFR reduced, reduced Na excretion

Liver
- Fatty liver (esp in kwashiorkor, a little in marasmus), decreased endogenous glucose production

Pancreas
- Decreased beta cell function and enzyme production

Hormonal
- Decreased thyroid function and cortical, growth hormone responses

Gut
- Atrophy, decreased nutrient absorption, chronic inflammation, microbiome changes

Skin
- Decreased barrier role, subcut fat, decreased insulation, decreased collagen/protein reserve
Immune function
- Greater vulnerability to infection
- Atypical () response to infection
- Atypical manifestations of infection
- Poorer outcomes from infection

CNS
- Relatively protected so worrying sign of late stage
- Lethargy, irritability, decreased appetite and interaction with environment

963
Q

What is key to test in a child with malnutrition in terms of phased treatment and why?

A

Appetite -> helps differentiate between complicated vs complicated and helps determine phase progression during treatment

964
Q

How should you approach infection in an acutely malnourished child?

A
  • Assume infection -> all get 1st line Abx eg co-trimoxazole
  • Sick or not improving -> 2nd line Abx eg ampicillin and gent
  • Very sick or worsening fast -> 3rd line Abx eg cef or 3rd gen cephalosporins
965
Q

How to approach dehydration in context of malnourished child?

A

Usual signs DO NOT APPLY:

May (or may not) indicate dehydration:
- Skin turgor (loss subcut fat?)
- Sunken eyes (loss retrorbital fat?)
- Dry mouth / eyes (atrophied glands?)

Following ARE USEFUL:
- Recent changes in above
- Input / output history (+charts)
? Urine or watery stool?
- Weight changes (use SAME accurate
scales!)
- (Thirst)
- (Conscious level)

TREAT WITH GREAT CARE!:
- Risk of heart failure / worsening
- Oral / NGT whenever possible
- Specially forumulated solutions
- Monitor response closely
- e.g ‘High dependency’ beds)
- reweigh (wt up but not too fast)
- vitals better? (pulse; rr; CRT)
- passing urine
- more alert
- PAUSE / SLOW FLUIDS IF WORSENING
➔ Prevention better (early feeds / ORS / breastfeeding)

966
Q

How should you approach feeding an acutely malnourished child?

A

Phase 1 = stabilisation
- Small volumes (total and per feed)
- Frequent feeds eg 2/3hrly
- Light/easy to digest and metabolise -> PO whenever possible
- May need NG tube if not tolerating PO

Transition phase

Phase 2 = 2 rehabilitation
- Large volumes (total and per feed)
- Less frequent
- Nutrient dense

Offer breast milk before feeds ie don’t stop it

967
Q

What is the role of sensory stimulation in children with acute malnutrition?

A
  • Direct effect on CNS
    • Weak, sick -> do not crawl or explore environment
    • Do not engage with adults
    • Poor homes

Treat
- As essential as Abx etc
- Environment stimulating
- Structured play 15-30min/day
- Engage with carer eg exercise

968
Q

Reasons for community management of acute malnutrition?

A

Good for patients
- Prefer home treatment
Decrease:
- Nosocomial infection
- Abx resistance (also population benefit)
- Hospital costs

Good for carers
- Time for other children and home duties
- Increased early presentation and less absconders

Good for staff
- Less pressure on ward based care

Good for communities
- Empower them, improve impact, begin to address root causes

969
Q

What are the WHO principles of inpatient management for malnutrition?

A
  1. Hypoglycaemia: IV/NG dextrose
  2. Hypothermia: skin-2-skin, warm fluids
  3. Dehydration + shock: ReSoMal (ORS), ?fluid
    bolus, transfuse if Hb<4
  4. Electrolytes - TNa IK: ReSoMal or F75
  5. Infection: all SAM → Abx, consider specific
    infections (malaria, worms, candiasis)
  6. Micronutrients: Vitamin A (eyes), Zn, Fe (but
    not early)
  7. Feeding:
    <6 months: encourage breastfeeding before
    formula
    >6 months: F75, monitor for appetite
  8. Catch-up growth: F100 2-3 days/plumpynut,
    fortified local foods
  9. Stimulate and support: play to encourage
    development
  10. Follow-up: immunisations, social work, nutrition

Remember: classical signs of dehydration do not
apply in malnutrition
Monitor UO, weight trend and level of
consciousness

NB:
MUAC will respond to treatment -> don’t always need weight
MUAC only relevant between 6 months to 5 years

970
Q

What are the complications of malnutrition?

A

Severe 3+ oedema
Unable to sit up
Hypothermia
Vomiting
Metabolic disturbance
Severe infection
Open skin lesions
Failed appetite test

971
Q

Long term consequences/associations of malnutrition?
Underlying causes of malnutrition?

A

Short-term (morbidity, mortality) vs long-term
(adult height, cognition, economic performance,
reproductive performance, obesity, CVS + metabolic disease)

First 1000 days of life issues

Underlying causes: household food security,
inadequate care + feeding practices, access to
healthcare + household environment
Basic causes: quantity + quality of resources (land,
education, employment, £$, technology),
financial/human/physical/social capital, and
social/economic/political context

972
Q

Causative organism and transmission for diphtheria?

A

Corynebacterium diptheriae
Aerobic, Gram-positive rods with club shaped
morphology

Transmission by respiratory droplets and close
contact and communicable for 2-6w without
antibiotics, 48h with antibiotics

973
Q

Pathophysiology of diphtheria?
Clinical symptoms?
Complications?

A

Patho:
Exo-toxin mediated disease
Death of mucosal epithelial cells > formation of grey
fibrinous pseudo-membranes
Lymphatic and haematogenous spread >
disseminated disease, shock and multiorgan
involvement

SYMPTOMS
2-5 day incubation period
Pharyngitis or laryngitis
Gross lymphadenopathy -> ”bull neck”
Adherent pseudomembranes
Respiratory failure due to airway obstruction or aspiration of pseudomembranes
Initially isolated spots of grey/white, coalesce to form membrane that bleeds on contact
Examine: uvular, palate, oropharynx
and nasopharynx
Simple: one area only
Extensive: >1 area

COMPLICATIONS
Myocarditis (2-7 weeks)
Neuropathy and paralysis (1-3 months)
Renal failure

974
Q

Treatment of diphtheria?

A

ANTITOXIN
Indications: tonsillar plaques, pseudomembranes,
shock or systemic signs
Contraindications: >7 days symptoms, unresponsive,
intractable shock or airway compromise
Risks: anaphylaxis (<1%), febrile reaction (4%), serum
sickness (8%)
Dose according to severity of disease

ABx
Severe: IM/IV benzylpenicillin or erythromycin
Mild-moderate: PO penicillin V, erythromycin or
azithromycin

VACCINATION
Vaccinate by age: Pentavalent (if 6w – 6y) or Td if
(>7y)

CLOSE CONTACTS
Prophylactic antibiotics: single dose IM benzathine
penicillin or azithromycin or erythromycin PO for 7 days
Vaccination by age

975
Q

What are some complications of neonatal tetanus?

A

Death (high mortality rate)
Small head circumference
Microencephaly
Other cognitive/developmental impacts eg hand to eye coordination tasks, lower summated developmental score
Neurological abnormalities
Behavioural problems

976
Q

How to prevent neonatal tetanus?

A
  1. Clean delivery practices
    - hands/delivery surface/cord care
  2. Tetanus toxoid protects both mother and child
    Un-immunised pregnant women:
    2 doses tetanus toxoid
    - 1st dose as early as possible in pregnancy
    - 2nd dose at least one month later/3 weeks before
    delivery.
  3. Infection does not provide natural immunity -> need to vaccinate
977
Q

Global progress in neonatal and maternal tetanus elimination since 2000?

A

Highest in Africa and South Asia

World Health Organization estimate shows is 25,000 deaths from NNT in 2018, which means an
88% reduction from since 2000.

Maternal tetanus and NNT have not yet been eliminated (July 2019) in 12 countries

978
Q

A 49M is a type 2 diabetic with poor control. He had a below knee amputation 7 days before for
chronic diabetic foot infection.
He has had 1 day feeling generally unwell, fevers.
Breathing difficulty
Not eating. Difficulty swallowing.
O/E Generally unkempt.
Stump was covered in dirt, wound opening up.
Day 1 admission:
Doctor called he couldn’t open his
mouth, drooling
Muscles were rigid, sweating profusely.
BP 200/160, HR 120, RR 36, Temp 39oC
Bloods: WCC 12, Neut 11, Hb 90, Plt 400

Ddx?

A

Tetanus
Strychnine poisoning
Narcotic withdrawal
Hypocalcaemic tetany
Neuroleptic malignant syndrome
Phenotiazine dystonic reaction
Rabies
Stiff-person syndrome

979
Q

What is the management of generalised tetanus?

A

Support
Airway -> Tracheostomy
Muscle spasm control
1st Benzodiazepines (Diazepam up to 20mg/kg/day)
2nd Chlorpromazine
3rd Vecuronium
Magnesium sulphate

Environment control
Minimise light, sounds, any stimulus that can cause
spasms

Turn off toxin
Neutralise toxin -> Human hyperimmune globulin or Anti-tetanus globulin (equine)
Source control -> surgical debridement
Antibiotic -> Metronidazole 500mg TDS IV 7-10 days

Complications
Autonomic dysfunction -> Labetolol or esmolol, Magnesium sulphate infusion*

Prevention
Give tetanus toxid vaccination -> No natural immunity derived from infection, therefore need immunisation course

980
Q

Tetanus vaccines
- what, schedule and who for?

A

DTaP/IPV/Hib/HepB then DTaP
4 in 1: 3 years and 4
6 in 1: 8, 12 and 16

Tdap for preteens
3 in 1: age 14

Td
Every 10 years

981
Q

Which of the following statements
about tetanus is true?
a) Caused by Gram-negative anaerobic bacilli
b) Neonatal tetanus elimination is defined as <1 per
10,000 live births in every district each year
c) Can only be diagnosed microbiologically
d) Human immunity to tetanus cannot be naturally-
acquired
e) Patients are always confused or unconscious

A

D

982
Q

What is neonatal and maternal tetanus elimination defined as?

A

Elimination defined as <1 NNT case per 1000 live births in every district per year

983
Q

Triad of clinical features for tetanus?

A

Trismus, ricus sardonicus, opisthotonos

984
Q

Which malnourished children need Vitamin A replacement?

A

Recent measles

Deficient

985
Q

Define outbreak

A

The occurrence of disease episodes in greater numbers than would be expected at a particular time and place

986
Q

Ddx for acute, suppurative, unilateral parotitis in children?

A

Acute bacterial infection eg staph aureus, strep pyogenes, Haemophilis influenzae

Neonates -> gram neg

HIV -> Strep pneumoniae

Endemic regions eg Laos/rice farming -> Burkholderia pseudomallei

If unvaccinated -> mumps (but not suppurative and becomes bilateral in majority of cases)

Other viruses:
CMV
EBV
Influenza A
Parainfluenza
Enteroviruses

987
Q

Epi of travel related conditions

A
  • Road traffic accidents account for 23% of injury related deaths PA globally
    Exacerbation/complications of chronic illness & motor vehicle accidents are the leading causes of death and severe disability in travellers
    >95% of travel associated illness is not vaccine preventable
988
Q

What questions should you ask a patient in travel medicine clinic?
What are the 5 interventions to consider?

A

○ Things to check:
▪ Age
▪ Comorbidities
▪ Risk threshold
▪ With whom
▪ Reason for travel
* What do they plan to do, where and with whom?
▪ Country
▪ Area
▪ Mode of travel
▪ Accommodation
▪ What is going on

  • How long are they going for, what is the season?
    ▪ Length of stay
    Seasonality
  1. Malaria prevention
  2. Vaccines
  3. Non-vaccine preventable infections
  4. Other: sexual health/altitude/DVT/Trauma/Security/Insurance
  5. Patient education
989
Q

What is the communication framework used in travel medicine?

A

DARN
Desire, ability, reasons, need
1. Ask permission to start agenda
2. What does patient think about their health
3. Ask questions to find out what they already know
4. Tell the patient information in a way that’s easy to understand
5. What is the patient’s understanding

990
Q
A

1st trimester
* Concern for mother
* Miscarriage
* Ectopic
* Bleeding
* Retained products
* N&V
* DVT
* Infection

2nd trimester
* Concern for mother
* Congenital abnormalities
* Anaemia
* DVT
* Gestational diabetes
* HTN
* Infection

3rd trimester
* Concern for mother and baby
* Premature labour
* Stillbirth
* Post-partum haemorrhage
* DVT
* HTN/Pre-eclampsia/Infection

991
Q

What test can you do on a pregnant women who comes to travel clinic and why?

A

Fetal USS to look for heartbeat
Once heartbeat is seen the risk of miscarriage<5%
Can also look for
- congenital abnormalities
- position of fetus
- fetal growth
- position of placenta

992
Q

What puts pregnant women at higher risk of blood clots when travelling?
How can you reduce this risk?
What vaccines can pregnant women be offered?

A
  • flights/journeys > 4hrs
  • Older age
  • previous clot
  • family history
  • additional clotting disorder
  • Obesity
  • severe oedema
  • Immobility
  • Other medical conditions

REDUCE VIA
* Compression stockings
* Hydration
* Mobilization
* Isle seat
* No ETOH, sleeping medication
* LMH safe

  • UK all get pertussis vaccine 16-32 weeks
  • RSV vaccine from 28 weeks (Aug 24 UK)
  • Flu & COVID recommended
  • No evidence harm from inactivated vaccines
  • Live; balance risk vs theoretical foetal harm
  • Inadvertent vaccine administration NEVER indication for termination
993
Q

Pros of inactivated vaccines and examples compared to live-attenuated vaccines?

A

INACTIVATED
Safe
Less robust immune response
Multiple doses often needed
Waning immunity
* Pneumococcus
* IFV
* Hepatitis
* D/T/P
* Typhoid

LIVE-ATTENUATED
Robust immune response
Often lifelong immunity
BUT
Potential vaccine derived infection
* MMR
* YF
* OPV
* Zoster

994
Q

Describe the timing of administration and immunological response to vaccine Ag?
What influences vaccine response?

A
  • Early administration
    ○ Mainly addresses GC-independent memory B-cells
  • Germinal-center activity persists for weeks to months resulting in more memory B-cells that can be addressed by revaccination
  • Long term protection
    ○ Ab persistence above threshold +/- memory cells
    Rapid clonal expansion on exposure to pathogen

RESPONSE
* Early Ag specific Ab
* Efficacy = avidity + specificity + neutralizing capacity
* Long term protection = persistence Ab + memory + rapid clonal expansion
Eg B cells and T cells

995
Q

What are some examples of primary vs secondary immunodeficiency?

A

Primary : Inherited, can affect all areas, usually severe e.g.
* SCID
* CVID
* Di George
* IgA deficiency

Secondary/acquired : Physiological state, illness or treatment for a condition that reduces your immune function e.g.
* HIV
* Extremes of age
* Pregnancy
* Malignancy
* Post BMT
Medication etc

996
Q

Who might need more vaccines due to attenuated response or increased risk of infection?

A
  • Asplenia
  • Complement deficiencies
  • Medication
  • HIV
  • Pregnancy
  • IVIG
  • Post BMT/CART-T
997
Q

In travel clinic what vaccinations would you consider for?
Person:
40-year-old born in India, moved to UK age 20
38-year-old born UK never traveled abroad
6-month-old well, vaccines to date
3-year-old well, vaccines to date
Place: Mumbai, India
Time: 6-weeks

A
  • Measles
    ○ Give mum MMR if not pregnant
    ○ 6 month year old -> give early MMR -> but does not count as initial dose of MMR -> needs full repeat when of age
  • Hepatitis A
    ○ Mum: might have had exposure, can test but can also just vaccinate
    ○ Dad: vaccinate
    ○ 3 year old: immunise due to public health management
    ○ 6mth old: no, if breast fed then no risk
  • Typhoid
  • Dengue
  • Rabies
998
Q

What issues do you have regarding Ocreluzemab and infection?

A

Ocreluzemab:
* B cell depleting
* Severely reduced clonal expansion
* Reduced efficacy
* Reduced safety
* Increased risk of infection, especially intracellular like pneumococcal and skin and soft tissue infections
* Increase risk of reactivation of infections, esp HBV so check liver function before starting treatment
* Vaccinate against pneumococcal disease prior to starting Ocreluzemab

999
Q

Management of animal bite eg monkey

A
  • Wash wound for 15mins with soap and water
  • Seek medical attention ASAP
  • DTP booster if required
  • May need Abx
  • Simian HerpesB: Macaque monkeys in SEA, Aciclovir within 5 days for 14 days
    ○ Rare but >85% mortality
  • Rabies PEP+/-HRIG
1000
Q

What vaccines does the following traveller need?

* 45 yr old photographer
* Work trip to New England USA
* Going in May for 4 weeks 
* Leaving in 3 weeks
* Splenectomy 2 years ago after road traffic accident in SA
* No additional medication, advice, vaccinations or follow up in UK Works for lonely planet -> mixed urban and rural work
A

Additional vaccinations
* Haemophilus influenza type b
* Meningococcal ACWY
* Meningococcal B vaccine
* Influenza
* Pneumococcal

Prophylactic/standby Abx
* PenV
* Consider stand by Abx

Other infection risks
* Malaria
* Babaesiosis

Give other advice:
* Trousers tucked into socks
* Permethrin spray
* Tick check and removal

1001
Q

What vaccines would you offer the following patient in travel clinic?
Person: 28 year old diabetic
Place: Hajj pilgrimage
Time: leaving in 2 weeks

A
  • Men ACWY at least 10 days prior to travel
  • Polio if travelling from a country with wild-type polio
  • Yellow fever if travelling from ‘at risk country’
  • Other things to consider:
    ○ COVID-19
    ○ Flu
    ○ Hep B
    ○ Rabies
    ○ MERS-CoV
    ▪ Close contact with animals eg camels, unpasteurised camel products
    ○ Diarrhoea
1002
Q

What do you need to consider when deciding where to build an Ebola treatment centre?
What are some specific considerations for the zones?
What are the three key water sources needed in ETC?
What staff are needed?

A

Roads for supply access
Water
- ?from where
- any nearby streams/bore holes?
- risk of viral contaminated into ground water/village water supply
Electricity (ideally mains)
Size and space (12 beds thought to need 2500metres squared)
Phone signal
Ground water level/flood plain -> ideally need at least 1.5 metres above ground water level
Need big buffer zone around ETC

If worried about contamination of water supply -> check Cl levels as remain in water, viral levels hard to determine

Buildings needed:
- ensure ancillary eg lab, pharmacy, morgue, staff areas

Ensure potential for holistic care: eg visiting, spiritual needs, children/neonates, survivors, dying

Donning and doffing
- ideal need mirrors, clocks (35/40mins before need to exit zone), lots of space, well lit
- need buddy check and third person
- doffing done by someone else based on instructions, need multiple lanes and hygienists

WATER
- 0.5% chlorine (200L per patient per day)
- 0.05% chlorine (150L per patient per day)
- drinking water (50L per patient per day)

Staff:
- hygienists
- laundry staff
- medics
- nurses
- logistics
- psychologists
- rota/record keepers

1003
Q

What are the two key principles for constructing outbreak treatment centres?

A
  1. Risk zoning (need No pathogen and pathogen zones, need whole centre to be separated from outside world)
  2. Flow -> need patient flow pathway and staff flow pathway which will be different. UNIDIRECTIONAL ONLY

Need protocols for case

1004
Q

What would a blood film of Bartonella bacilliformis look like?

A

Gram negative aerobic, pleomorphic, flagellated, motile, coccobacillary
INTRACELLULAR

1005
Q

How does Pellagra present? What causes it?

A

Pyoderma
Dermatitis
Dementia

Vit B3 deficiency

1006
Q

What are the skin features of scurvy?

A

Perifollicular haemorrhages

1007
Q

Elapid envenoming features

A

Neurotoxicity, ptosis, breathing difficulties (eg mambas, cobras, kraits)

  • they usually warn you before they bite, ie they rear up
1008
Q

Where are Russell’s vipers found?

A

Asia
NOT Africa

1009
Q

What is the most common cause of bacterial meningitis in Africa (not in meningitis belt)?
How can you differentiate bacterial from TBM or cryptococcal disease?
What are other potential causative organisms of bacterial meningitis?
What is the suggested treatment and duration?

A

Streptococcus pneumoniae

Bacterial will occur in hours to days, TBM/Crypto will be days to weeks

Staphylococcus aureus
Streptococcus pyogenes
Haemophilus influenzae
Non-typhoidal salmonella
Salmonella typhi
Escherichia coli
If pregnant - ?Listeria monocytogenes

Apart from Listeria, treat with 3rd generation cephalosporin, ideally 14 days but evidence mixed -> also check local resistance patterns

1010
Q

Ddx for generalised, non-pruritic rash with small umbilicated papules?

A

HIV
EBV
CMV
Cutaneous TB
Non tuberculosis mycobacterial infections
Mollusca contagiosum
Melioidosis
Bartonella spp. (causing bacillary angiomatosis in severely immunocompromised patients -> usually erythematous lesions)
Histoplasmosis
Talaromycosis
Cryptococcosis

Lymphomas
HHV-8 castleman’s disease

1011
Q

Key very basic presenting features for EBV/CMV

A

Infectious mononucleosis
Hepatosplenomegaly
Lymphadenopathy

1012
Q

Triad of features for acute fascioliasis?

A

Fever
Eosinophilia
Elevated liver enzymes

1013
Q

A 43 year old man presents after a trip one month ago to Mozambique with high fever, eosinophilia, a cough, headache and some diarrhoea. He did not take malaria chemoprophylaxis and reports some freshwater contact in a small lake near Maputo. What are the two key diagnoses to rule out?
What investigations would you perform for these and issues with their sensitivity/specificity?

A

Malaria -> blood films
Katayama syndrome
- need urine and stool for Ova and cysts of Schistosoma but these may not appear until 3 months after infection and patients can be infected with different stages of schistosomulae at once

1014
Q

What is Katayama syndrome? How does it present and how is it managed?

A

Acute schistosomiasis -> acute hypersensitivity reaction thought to be result of initial egg deposition by adult trematodes

Incubation period: 2-12 weeks

Fever + urticaria + dry cough +/- dry wheeze

CXR: patchy pulmonary infiltrates or micronodular changes in the lower lungs

NB: eosinophilia may be delayed, ie not seen after a couple of weeks. In addition, may need to repeat urine/stool tests for ova as may not appear for at least 3 months. Can do PCR if available.

Manage with praziquantel after adults have developed as it is not effective against immature flukes. -> may need repeat courses due to infection with many schistosomulae.

1015
Q

Name some diseases caused by flaviviruses and their relevant vectors?
Where are they found?
When do the vectors bite?

A

Dengue -> aedes aegypti or albopictus -> Asia>Latin America>Africa -> DAY

Yellow fever -> aedes -> Africa + LA -> DAY

Zika -> aedes -> LA>Caribbean>few
African/Asian cases -> DAY

Japanese encephalitis -> culex -> Asia -> dawn/dusk/night

1016
Q

What type of virus is Chikungunya caused by and vector?
Where is it found?
When do the vectors bite?

A

Alphavirus
Aedes
LA,Africa,Asia
DAY

1017
Q

Features of yellow fever?
Life cycle?
What vaccine exists and pros and cons?

A

Yellow fever -> aedes -> Africa + LA -> DAY
Positive single-stranded RNA virus

Incubation: 3-6 days, max 15

Life cycle:
urban - aedes aegypti biting infected humans then uninfected and circulates
sylvatic - aedes africanus biting infected non-human primates and circulates
intermediate/savanna - aedes biting non-human primates then humans and circulates

  • Most asymptomatic
  • Fever, headache, chills, muscle pain, nausea
  • Most self-limiting
  • 1 week
  • 10-25% haemorrhagic manifestation, kidney and liver damage +/- encephalitis
  • Case fatality rate of 20-50%

Concern regarding its spread as can go with aedes which are found almost everywhere

VACCINE - live attenuated
Safe
✓Massively used since 1938
✓Rare complications
Efficient
✓Fast neutralizing immune response
✓One dose; long-lasting protection - now thought life-long

CONs
Production
✓Need for embryonated egg
✓Q/C & batch release time
Storage & transport
✓Need for strict cold-chain

1018
Q

What are the 3 serious adverse events associated with yellow fever vaccination?

A

Hypersensitivity reaction
1:100.000 doses

Vaccine-associated viscerotropic disease (YEL-AVD)
1: 200.000-300.000 doses
1: 40.000-50.000 doses >= 60 years old

Vaccine-associated neurotropic disease
1: 150.000-250.000 doses

Revaccination has not been associated with serious adverse events.

1019
Q

Contradictions to yellow fever vaccine and patients to be cautious with it in?
Who might you give a booster to?

A

CIs:
* < age 6 months.
* Persons with a confirmed anaphylactic reaction to a previous dose of YF vaccine.
* Persons with a confirmed anaphylactic reaction to any of the components of the vaccine, including egg
* Persons with a history of thymus disorder* or removal of the thymus gland for any reason
(thymectomy) including incidental thymectomy (e.g. during cardiac surgery).
* Persons with primary or acquired immunodeficiency including symptomatic HIV infection and
asymptomatic HIV infection when accompanied by evidence of impaired immune function
* Persons who are immunosuppressed
* Persons with a first-degree** family history of YEL-AVD or YEL-AND following vaccination that was not
related to a known medical risk factor (in case of an unidentified genetic predisposition).
* Individuals aged 60 years and older travelling to countries and areas designated by WHO as areas with
low potential for exposure to yellow fever virus (see section on people aged 60 years or older).

Precautions:
* Infants aged six to eight months
* Individuals aged 60 years and older
* Breastfeeding women
* Pregnant women
* Those taking low dose steroid or non-biological oral immune modulating drugs
* Individuals living with HIV (with a CD4 count greater than 200 and a suppressed viral load)

BOOSTER (consider if)
- If person is working/living in endemic region or with the virus ie high risk setting and last had vaccine over 10 years ago
- If person had original vaccine over 10 years ago and in a slightly immunocompromised state, eg <2 years old, when pregnant, before undergoing BM transplant, when infected with HIV or when immunosuppressed

1020
Q

What is the Public health strategy in endemic areas for yellow fever?
What do you do if you are low on vaccine supply?

A

Protect populations at risk
- Mass vaccination campaigns (need 80+% in at risk areas to prevent outbreaks)
- Catch up vaccination campaigns
- Vaccinate every child
- Evaluate risk to prioritise resources

WHO recommendation 2016: YF fractional dose vaccination should be considered in response to an emergency situation in which current vaccine supply is insufficient.

Prevent international spread
- Apply the IHR: Monitoring and surveillance, strengthening public health capacity, collaboration and communication, rapid response and containment
- Build resilient urban centres

Contain outbreaks rapidly
- Detect early: strengthen lab and surveillance capacity
- Timely and equitable access to vaccine supply
- Respond immediately

1021
Q

What are the international health regulations? (IHR)

A

International Health Regulations (IHR), a legally binding framework established by the World Health Organization (WHO) to strengthen global health security. The IHR (2005) aims to help countries prevent, detect, and respond to public health risks that have the potential to spread internationally, ensuring that outbreaks and other health emergencies are managed effectively to minimize their impact.

Involve:
Monitoring and surveillance, strengthening public health capacity, collaboration and communication, rapid response and containment

1022
Q

What are the international health regulations?

A

International Health Regulations (IHR), a legally binding framework established by the World Health Organization (WHO) to strengthen global health security. The IHR (2005) aims to help countries prevent, detect, and respond to public health risks that have the potential to spread internationally, ensuring that outbreaks and other health emergencies are managed effectively to minimize their impact.

Involve:
Monitoring and surveillance, strengthening public health capacity, collaboration and communication, rapid response and containment

1023
Q

Who does the DENGVACIA vaccine work in? Who is it dangerous to give to and why?

A

Efficacious and safe in SEROPOSITIVE persons: 72-80% against dengue of any severity; >90% against severe dengue

DO NOT GIVE TO SERONEGATIVE patients -> increases risk of severe dengue

1024
Q

What licensed vaccines are available for dengue?

A

Dengvaxia
- 3 doses over 12 months
- check serostatus before giving
- 9-45 year olds

TAK003/Qdenga (Takeda)
- 2 doses (0 and 3 months)
- WHO: 6-16 years in endemic settings

1025
Q

Should you vaccinate travellers for dengue? What other advice might you give?

A

Only if they are seropositive (you need to test them)/have had dengue in past, aged 4+ and are travelling to high risk areas, areas with ongoing outbreaks or will come into contact with virus through work

Day time biting so use repellant
Risk highest during and just after rainy season

1026
Q

How is Zika transmitted?
How does Zika present?

A

Aedes mosquito bite
Sexual
Blood transfusion
Pregnancy

S/S:
- Asymptomatic 50-80%
- Mild disease 20-50%
- Complications in <1%

1027
Q

What are some of the severe complications with zika infection?

A

Neurologic eg GB syndrome, transverse myelitis, meningoencephalitis
Ocular
Thrombocytopenic purpura
Transient myocarditis
Highest risk of birth defects during 1>2>3 trimester

In 20-30% of babies who are infected:
Fetal loss
Congenital zika with microencephaly
Asymptomatic infection including sequelae in baby/child of:
* Seizures
* Hearing loss
* Visual impairment
* Dysphagia
* Development delay

1028
Q

What advice should be given to couples regarding the sexual transmission of zika?

A

If male travelling and partner pregnant OR female pregnant and travelling with male then barrier methods for whole pregnancy

If female traveller pregnant but male did not travel -> barrier methods for 2 months after last possible exposure

If male traveller and partner NOT pregnant OR female traveller not pregnant but male partner also traveller -> avoid conception + barrier methods for 3 months

If female traveller not pregnant and male partner did not travel then avoid conception and barrier for 2 months after Zika exposure

1029
Q

Features of Chikungunya
- Virus
- Incubation
- Signs/symptoms

A

Alphavirus, genus Togaviridae

Incubation: 2-6 days

90% of patients will have:
fever (up to 1 week), myalgia (up to 10 days), polyarthralgia, polyarthritis or both (weeks to months)
Rash for 1 week in 50% patients

Viraemia will last 5-7 days
IgM 3-8days after onset to up to 3 months
IgG 4-10 days after onset, persists for years

1030
Q
A

1 Maternal risk |2 Antenatal risk |3 Perinatal complications

Dengue:
1 Increased risk of severe disease and death; more difficult clinical mx of severe dengue
2 Increased fetal losses in the first half of pregnancy (data from multiple cohorts, substantiated by a meta-
analysis)
3 Severe neonatal infection with sepsis-like symptoms and acute respiratory distress reported in case report: most other cases mild

Zika
1 No increased risk, prolonged viremia
2 Congenital Zika Syndrome
3 No increased risk

Chikungunya
1 No increased risk except for pre-term labour
2 Miscarriages documented in three (2%) of 678 participants in one study and no increase in number of stillbirths, prematurity, or malformation
3 Documented transmission rate of 28–49% with severe neonatal infection (encephalopathy) in 53% of newborns in
one study; severe neonatal infection with encephalopathy shown in four studies

1030
Q

Who would you give a Chikungunya vaccine to?

A
  • Recommended for:
    ○ 18+ yr olds and travelling to country with outbreak
  • Consider in:
    ○ Travelling to non-outbreak place but evidence of chikungunya virus transmission among humans within the last 5 years in people who
    ▪ Aged >65 years old, esp with medical conditions who are likely to have moderate exposure OR
    Ppl staying for cumulative period of 6 months or more
1031
Q

Basic facts on Oropouche virus

A
  • Genus Orthobunyavirus, Peribunyaviridae family
  • Transmitted by midges (Culicoides paraensis) and
    mosquitoes (Aedes spp)
  • Reservoir pale-throated sloths, non-human
    primates, and birds
  • Most infection are mild (symptoms similar to
    dengue)
  • Approx 4% develop neuroinvasive disease
  • Report of vertical transmission!

Rising cases in Latin America eg Brazil

1032
Q

Case fatality rate definition?

A

The probability that a case will die from the infection being studied

1033
Q

What is selection bias? What are some types of it?

A

an error in selecting the study population such that either the people who are selected to participate are not representative of the population of interest or the comparison groups are not comparable

Survivorship bias
Sampling bias
Attrition bias
Non-response bias
Volunteer bias
Undercoverage bias

1034
Q

What is information bias? Why does it occur and what are some types of it?

A

Any error in the measurement of exposure or outcome resulting in systematic differences in the accuracy of information collected between groups being compared

Occurs
- In case-control study: if accuracy of exposure data differs between case and controls
- In cohort study: if accuracy of outcome data differs between exposed and unexposed groups

Recall bias
Observer bias
Performance

1035
Q

Top 5 leading causes of global under 5 mortality?

A
  • Neonatal disorders
  • Pneumonia
  • Diarrhoeal diseases
  • Congenital defects
  • Malaria
1036
Q

Features and management of severe pneumonia in children

A

Cough + dyspnoea + danger signs:
Severe respiratory distress:
grunting, very severe chest
indrawing
· O2 sats <90% in air or
central cyanosis
· Inability to breastfeed/drink
· Reduced
LOC/lethargy/convulsions

MANAGEMENT
Admit
· O2 therapy if known low sats,
central cyanosis or severe resp.
distress
· IV antibiotics (local spectrum or
IV amp and gent if unknown)
· Care with fluid balance
· Anti-pyretics if child distressed,
bronchodilators if wheeze

1037
Q

Key causes of LRTI in children? (PERCH study of pneumonia etiology in 7
countries across Africa and Asia)

A

71% of all LRTI due to 10 organisms:
RSV, flu, metapneumovirus, rhinovirus, paraflu, s.pneumo, s,aureus, h.influenza, TB, PCP

1038
Q

What did the CHAMPS post-mortem study in SSA and S. Asia show with regard to LRTIs?

A

High levels of Gram –ve organisms (eg Klebsiella, non-typhoidal haemophilus influenzae E Coli) and
Strep pneumonia, in fatal LRTIs

1039
Q

Methods of delivery of oxygen

A

– Cylinders: produced in manufacturing plants. Need regular refilling. Often means
intermittent supply
– Concentrators: extract nitrogen from air in the atmosphere to leave O2. Done at the
bedside. Can supply up to 5 patients with flow-splitters. Need constant electricity. Need
cylinders as back-up in case of power-cuts
– Central piped O2: Piped from large O2 source e.g. concentrator. High cost, usually only in central hospitals.

1040
Q

When can you stop oxygen therapy in a child?

A

Can stop if O2 can be disconnected for 10-15mins with no emergency signs or SpO2>90% in air.
Monitor for 24hrs as tends to worsen overnight

1041
Q

What are the methods of administration of oxygen and flow rates in kids?

A

Nasal prongs, face-mask, head-box/tent, nasal catheter
Nasal prongs recommended as provide ~5cm PEEP to help improve oxygenation and
most efficient
Flow rates: 0.5-1L/min neonates (<1mth)
1-2L/min infants (<1yr)
1-4L/min pre-school (<5yr)
1-6L/min older children
Check for mucous plugging and position 3hourly
Humidification needed for flow-rates >4L/min

1042
Q

What features in children would make you prioritise them for oxygen therapy?

A

Central Cyanosis (bluish discoloration)
Decreased consciousness
Grunting with every breath
Nasal flaring
Severe palmar or conjunctival pallor
Acute coma or convulsions >15mins
Severe chest indrawing
Tachypnea (rapid breathing by age) >70/min
Head nodding

1043
Q

What did the COAST trial show?*

A
1044
Q

TB screening for children with symptoms?

A

1) Do TB symptom screen
If positive:
2) Do CXR + molecular diagnostics eg rapid diagnostic test like Xpert MTB/RIF, Xpert Ultra, LAMP or Truenat

Samples from:
- Sputum/induced sputum
- Early morning GA
- Stool
- NPA
- Biopsy samples e.g. of LN
- CSF if LP

1045
Q

What did the SHINE study show?*

A

Children with non-TB could be treated for 4 months rather than longer standard course of treatment

1046
Q

How is non-severe TB defined and how should it be treated in children?

A

‘Nonsevere’ =
- non-cavitatory, non-miliary
pulmonary TB in one lung
lobe
- Peripheral LN TB
- Intra-thoracic LN TB
without airway obstruction
- Simple TB pleural effusion

Regime =
2 months HRZ(E)
2 months HR

1047
Q

How should contacts of TB positive child be treated?

A

All asymptomatic household
contacts should be offered TPT

Children <5 years and PLHIV
particularly high risk => target
populations for TPT

TPT options:
6 months isoniazid
3 months isoniazid and rifampicin
4 months rifampicin
3 months weekly rifabutin and
isoniazid
1 month daily rifabutin and
isoniazid

1048
Q

What can children with suspected MDR TB be treated with and based on what trials?*

A

6 month BDLLfxC can be used in all (that have not had >1 month exposure previously to Bdq, Dlm or Lzd)

BEAT-TB and endTB trials

1049
Q

TB CHAMP AND V-QUIN

A
1050
Q

When do most childhood deaths occur?
Related to this, what are the key causes?

A

50% of under 5 mortality in 1st month of life – most in the first week
* C.1/3rd infections, 1/3rd birth complications, 1/3rd prematurity

1051
Q

WHO indicators of Serious Bacterial Infection in neonate?

A
  • Fast breathing (respiratory rate ≥60 breaths/minute
  • Severe chest in-drawing
  • Fever (temp ≥38˚C)
  • Hypothermia (temp <35.5˚C)
  • No movement at all or movement only on stimulation
  • Feeding poorly or not feeding at all
  • Convulsions

very poor specificity

1052
Q

How should you investigate and manage an unwell neonate (particularly in low resource setting)?

A

Investigations: Blood culture + LP if safe, malaria RDT, BM if available

A: support if needed (neutral head position, chin lift)

B: O2 (0.5-1L/min) via nasal prongs if seizing/resp distress and available

C: NG feeding EBM if not breastfeeding

D: Treat low BM (2-5ml/kg 10% dextrose IV or NG)
Phenobarbitone for seizures (then phenytoin/benzos if needed)

E: Keep warm – hat, KMC
Phototherapy for jaundice if available – if not then transfer. If not possible, sunlight.
can influence methyl

IV antibiotics:
Go on local resistance pattern/local guidelines if available
If unknown Amp & gent
If meningitis concerns add 3rd gen cephalosporin

- Ceftriaxone difficult in small baby as displaces bilirubin so makes jaundice worse, cefotaxime better
1053
Q

What are some possibilities for pOPAT in infants 0-59 days with SBI where referral is not possible?

A

Hospitalisation is still the priority in these cases
* If not possible then if signs of critical illness:
– BD IM ampicillin or benpen, and OD IM gentamicin
– OD IM ceftriaxone or procaine penicillin, and OD IM gentamicin
“until hospitalisation is possible, up to 7 days”

  • If severe but not critical illness:
    – PO amoxicillin BD for 7d with IM gentamicin OD for 7d
    – PO amoxicillin BD for 7d with IM gentamicin OD for 2d
  • If fast breathing as only sign of pSBI then oral amoxicillin
    only BD for 7 days is acceptable
1054
Q

3 questions to ask about a child with severe watery diarrhoea?

A
  • Are they severely malnourished?
  • Are they shocked?
  • How dehydrated is the child?
1055
Q

Criteria for some vs severe dehydration in children in context of acute watery diarrhoea?
Management for each group?

A

SOME
2 or more of:
Restless/irritable
Sunken eyes
Thirsty/drinks eagerly
Skin pinch goes back slowly

MANAGE
Treatment Plan B (treat at 7.5% fluid deficit):
* Oral rehydration in hospital
over 4 hours
* Home if tolerates
* Zinc PO

SEVERE
2 or more of:
Lethargy or unconsciousness
Sunken eyes
Unable to drink or drinks poorly
Skin pinch goes back v. slowly (≥2secs)

MANAGE
Treatment Plan C (manage as 10% fluid deficit):
* IV or NG rehydration
(30ml/kg over 30mins-1hr then
70ml/kg over 2.5-5hrs + 5ml/kg/hr ORS
orally when tolerates)
* Hospital admission
* Zinc PO

1056
Q

Management of no dehydration in acute watery diarrhoea case in a child?

A

Means signs not meeting some/severe dehydration criteria

Treatment Plan A:
* Oral rehydration at home
50-100ml/loose stool <1
100-200ml/loose stool >1
* Zinc PO

1057
Q

What did the GEMs study show with respect to acute watery diarrhoea?

A

Top 5 pathogens for AWD = Rotavirus, shigella, ST-ETEC, cryptosporidium and
enteropathogenic E.coli

  • More recent studies suggest rotavirus still leading cause despite roll-out of
    rotavirus vaccine
1058
Q

What are the two drip rates in children for fluids vs blood per ml?

A

~ 20 drops/ml clear fluids
~ 15 drops/ml blood

1059
Q

General principles of diarrhoea and vomiting management in children?

A
  1. Replace lost fluid and give extra for ongoing loses
  2. Feed early and extra (only with-hold food in initial fluid replacement phase)
  3. Zinc PO (though limited evidence)
  4. Advise when to return (worsening/fever/blood in stool/persistent diarrhoea)
1060
Q

HIV testing in infancy? *

A
1061
Q

What is the package of care for HIV and starting ARVs in a child?

A
  • Screen for TB and other opportunistic infections and test/treat if present
    – delay ARVs for as short a time as possible (≤2-8 weeks) following TB/OI treatment
    commencement
  • Start ARVs according to national guidelines
  • Daily co-trimoxazole prophylaxis for all children <1 years (+ older children if CD4 <25%/200, or WHO stage 2,3,4)
  • TPT if TB contact and not concerned about active TB disease
  • Baseline bloods and CD4 count for monitoring purposes
  • Support parents and family
1062
Q

Does breast feeding increase risk of HIV transmission?

A
  • Undetectable does not mean untransmissible
  • PROMISE study
    with effective maternal ART, the risk of HIV transmission through breastfeeding could be minimized but not eliminated: 1% of kids still got HIV
1063
Q

What are the reasons for a positive RDT malaria test in child with recent malaria treatment?

How should you approach a recently treated child with a positive RDT?

A
  • ? Reinfection
  • ? Treatment failure
  • ? Persistent antigen but no malaria (if using PfHRP2) - roughly 4 weeks

Need microscopy or LDH-based RDT

If current infection confirmed WHO suggests:
* < 4 weeks from previous infection – assume treatment failure, give 2nd line anti-malarials
* If > 4 weeks from previous infection – treat as new infection but consider different oral agent if
available
* If severely unwell and delay in getting microscopy/LDH RDT treat as severe
malaria and other causes e.g. sepsis until results known

1064
Q

What is the best clinical test to confirm cerebral malaria?

A

Retinal fundoscopy
1. Patchy retinal whitening
2. Focal changes of vessel colour
3. Retinal haemorrhages - often white
centred +/- Papilloedema

1065
Q

What did the FEAST trial in paediatrics show?

A

Investigated fluid boluses in African children with severe infection, split children into no hypotension and hypotension groups
Trialled in each group, 3 sub-groups:
- saline bolus
- 5% Albumin bolus
- maintenance fluids only
(maintenance group not for hypotensive kids only non)
3.3% greater risk of 48hr mortality with any bolus given (p=0.003)

1066
Q

What are some associated health conditions with childhood sexual violence?

A

HIV/STDs
Pregnancy complications
Unwanted pregnancy
Alcohol use
Suicidal ideation
Depression
Attempted suicide
Difficulty sleeping
Cigarette use

1067
Q

Findings on examination of suspected victim of childhood sexual violence?

A

In most confirmed abuse cases examination will be normal. Some other findings include:
- Hymenal bumps, ridges and tags
- v-shaped notches (often superior / lateral) not extending to the base of the hymen
- vulvovaginitis
- labial agglutination
- vaginal discharge
- acute abrasions, lacerations or bruising of the labia, perihymenal tissues, penis, scrotum, or perineum.
- hymenal notch / cleft extending through more than 50% of the width of the hymenal rim, usually in 4 o’clock to 8 o’clock
- scarring or fresh laceration of the posterior fourchette
- presence of sperm confirmed on microscopy
- presence of sexually transmitted disease

1068
Q

Actions to take in case of suspected sexual violence in child?

A

History (patient led, with parent then ideally child alone)
Examination

  1. Record the findings
  2. Report for court/police
  3. Referral/liaison with social welfare / counselling / police etc
  4. PEP if <72 hours (eg in Malawi ABC/3TC + DTG < 30kg, TDF/3TC/DTG >30kg)
  5. STI prophylaxis (eg Malawi – metronidazole, erythromycin, gent, benzathine pen)
  6. Emergency contraception <5 days (eg in Malawi Levonorgestrel 1500mcg STAT)
1069
Q

Public health/policies that support developments in children aged 1-59 months (general concepts)?

Areas where intervention could be maximised?

A
  • Multiple disease-specific and integrated
    interventions e.g. HIV/malaria prevention &
    treatment, expanded vaccination, IMCI
  • Investment in health systems: free/subsidised healthcare for mothers and young children, training, CHW for prevention and basic care, infrastructure,
    equipment, supply chains, quality standards, data availability and use
  • Cross-sectoral collaboration: addressing
    malnutrition, water and sanitation, income
  • Governance: Sustained, high-level political
    commitment, stakeholder involvement, partner coordination
  • Focus on prevention: This age group is ideal for health
    interventions due to increased independence and lifestyle habit
    development
  • Scheduled well child visits: Ensuring scheduled health
    contacts for older children is key, as they are less likely to seek
    care when critically ill.
  • Interventions with potential: Priority interventions for children
    aged 5-9 include school feeding, micronutrient supplementation,
    health education, vision screening, deworming, mass drug
    administration for trachoma, vaccine catch-up, and mental health
    support.
  • Delivery platforms: Primary care services + outreach, schools
    and childcare centres, home and community platforms, child and
    social protection services, digital platforms
1070
Q

What are the common and rising causes of mortality in adolescents aged 10-14 years old? How is this split between male and female?

A
  • Infectious Diseases Prevail: Malaria, diarrhoea, and respiratory infections remain
    leading causes of death.
  • Emerging Challenges: Deaths from neoplasms (cancers), traffic accidents, and other injuries are on the rise.
  • Females: Significant burden of maternal
    causes
  • Males: High burden of injuries, particularly
    road traffic accidents and interpersonal
    violence.
  • Substantial burden of NCDs.
  • Regional variation -> high burden of HIV and TB in Africa.
1071
Q

Strategies to take for the management of global adolescent health?

A

Integrated approach: NCDs, mental health, injuries, SRH including maternal care, nutrition,
communicable disease and wider determinants
Health systems: workforce training to operationalize adolescent friendly services, sufficient
space, peer groups, communications campaigns, routine well-child visits etc.
Multisector e.g. road safety, school health, marketing regulation
* Supply: adolescent health included within paediatric training, centres for adolescent services
* Engagement and awareness: communications campaigns, teen clubs, adolescent involvement in
policy working groups
* Regulation: ban on tobacco advertising
* Wider sectors and platforms: developing school health
* Further progress: needs political commitment and funding for scale up and comprehensive
provision, and integration with social welfare workforce

1072
Q

Describe the altered pharmacokinetics of pregnancy

A
  1. Absorption
    ○ Reduced gut motility
    ○ Increased lung function
    ○ Increased skin blood circulation
  2. Distribution
    ○ Increased plasma volume, body water and fat deposition affecting volume of distribution (increased circulating volume to around 8L) -> consider eg with ARVs
    ○ Protein binding -> decreased plasma proteins
    (interpreting drug concs more difficult as more/less free unbound drug, more or less of a resulting effect)
  3. Metabolism
    ○ Hepatic enzyme activity could be increased (maternal hormones)
  4. Excretion
    ○ Increased glomerular filtration rate
    ○ Gastric motility reduced
1073
Q

What Abx should be avoided during pregnancy and which ones should you worry about?

A

AVOID
Tetracyclines (doxy -> staining, chelating in growing bones and teeth)
Streptomycin

Debatable
Fluoroquinolones
Aminoglycosides
Co-amoxiclav
Trimethoprim (folate agonist, neonatal haemolytic)

Other
Nitrofurantoin -> avoid at term
Metronidazole -> avoid high dose

1074
Q

What factors influence how much medicine reaches baby through breast milk?
How to do you minimise exposure?

A
  • blood level of medicine in mother
  • characteristics of medicine
  • amount of medicine passed into breast milk
  • amount of milk taken by baby per feed

Minimise exposure via:
* Withhold or delay therapy if possible
* Use a drug with poor penetration into milk
* Use an alternative route of administration
* Avoid nursing at peak drug concentrations
* Give drug before infant’s longest sleep
* As a last resort- ‘Pump & dump’ milk if short courses or discontinue breastfeeding

1075
Q

How do you manage pulmonary TB in pregnancy?

A

Risk of TB to mother and child outweighs risk of treatment
* First-line anti TB drugs cross the placenta but do not appear to be harmful
* Second-line anti TB drugs more risky. Avoid streptomycin (ototoxic to fetus)
* Continue breastfeeding (low concentrations of anti-TB drugs present)

  • Avoid streptomycin (and other 2nd line agents) if possible
  • Safety of pyrazinamide in fetus unknown
1076
Q

What is intermittent treatment of malaria in pregnancy and when should it be given?

A

IPTP

Sulfadoxine-pyrimethamine
- Start as early as possible in second trimester NOT before week 13
- give doses minimum of 1 month apart
- give 3 doses in total

SP is CI in first trimester of pregnancy as anti-folate

1077
Q

How should women in the first trimester of pregnancy with uncomplicated falciparum be treated?

A

Artemether-lumefantrine

1078
Q

How should pregnant women with severe malaria be treated?

A

Same as non pregnant
at least 24hr artesunate IV until they can tolerate oral medication (3 days ACT)

1079
Q

In the treatment of malaria in pregnant women, what drugs should be avoided?

A
  • Avoid primaquine
  • Avoid doxycycline
1080
Q

What are the pharmacokinetic factors affecting drug delivery in children and adolescents?

A

Absorption
* Administration route?
* pH
* Site of absorption
Distribution
* Lipophilicity vs hydrophylisity
* Volume of distribution
* Protein binding
Metabolism
* Hepatic maturity
* Microsomal enzyme processes
* Glucuronidation/sulphation
Excretion
* Renal function
* Gastric motility/peristalsis

1081
Q

Typical screening for children/adults from migrant communities in clinic?

A

Asymptomatic screening
Stool
- Ova, cysts & parasites microscopy
- Giardia PCR
Bloods
- FBC – anaemia, eosinophilia
- LFTs – in case of need TB treatment
- Vitamin D
- Serology – HIV / hep B / hep C / syphilis
- Schistosomiasis serology
- Strongyloides serology
Urine
- NAAT – chlamydia and gonococcus
(usually IGRA too)

1082
Q

What is PVL staph? What are the risk factors for getting it?

A

Panton-Valentine leukocidin = cytotoxin
PVL-SA is highly transmissible, lyses human neutrophils, and may cause recurrent skin and soft tissue infections (SSTIs) despite antibiotic treatment

Risk factors:
Close contact (eg, contact sports, military recruits)
Contaminated items
Crowding
Cleanliness
Cuts and compromised skin integrity.

1083
Q

Best tests for Bartonella?

A

PCR
IgG (IgM is very unreliable)

Diagnosis: serology (IgG) most useful with convalescent serology, PCR, culture, histopathology

LN biopsy best for PCR, also if you want tissue for PCR needs to be fresh

1084
Q

Features of Bartonella?

A

Facultative intracellular bacteria
Bite/scratch of infected cat or kittens, exposure to cat fleas, infected cats

Globally distributed, most commonly disease of children /young adults

Incubation: 3-10 days from exposure to primary inoculation lesion, 7-60 (14) for lymphadenopathy

Clinical manifestations:
- 85-90% localised cutaneous /LN infection
- PUO
- Disseminated disease - liver, spleen, eye, CNS (more common in children)

Complicaitons
- neuroretinitis
- encephalopathy, transverse myeltiis, cerebellar ataxia, radiculitis
- MSK including osteomyelitis
- endocarditis

Diagnosis: serology (IgG) most useful with convalescent serology, PCR, culture, histopathology

Treatment:
Localised disease -> sel resolving or 5 days azithromycin

Disseminated: Rifampicin + azithromycin, doxy + azithromycin for neuroreitinitis

If you are worried about a child -> can give doxy but ideally short course

1085
Q

Features of Mpox
- transmission
- incubation
- clinical manifestations
- complications
- diagnosis
- treatment
- vaccine

A

Orthopox virus - 2 clades I and II each with 2 subclades (a and b)

Transmission: close contact with lesions, resp droplets (prolonged f2f contact) and fomites, sexual

Incubation: 5-21 days (7), infectious 4 days prior to symptoms and until all lesions epithelised

Clinical manifestations:
- fever, flu like, lymphadenopathy
- centrifugal maculopapular rash after 3 days, spreads all over body including palm and soles, vesicles and pustules

Complications:
encephalitis
pneumonitis
conjunctivitis
secondary bacterial infections
CRF: 0-11% (endemic areas)
pregnant women -> fetal loss

Diagnosis: PCR from vesicle fluid, respiratory samples, blood

Treatment:
- supportive
- tecoviromat
- brincidofovir
- cidofovir

Vaccine:
- 3rd gen vaccine MVB-BA not currently licensed for pregnant women or children 3

1086
Q

Investigations for congenital syphilis?

A

Take paired maternal and infant syphilis serology
LP including PCR and VDRL
PCR on CSF, lesions, secretions
Neuroimaging - ?bony abnormalities
Audiology
Ophthalmology review
HIV/Hep B/ Hep C
STI screening (and treatment for parents)

Other
- anaemia, thrombocytopenia, abnormal WCC
- deranged LFTs
- CXR
- serology

1087
Q

Treatment for congenital syphilis

A

Procaine penicillin or benzyl penicillin
10 day course if symptoms or abnormal investigations
Single dose if well -> all normal Ix including CSF
if missed doses then restart treatment from day 1

Also treat parents

1088
Q

Two key adolescent assessments

A
  • HEEADSSS
    ○ Home
    ○ Education
    ○ Employment
    ○ Eating
    ○ Activities
    ○ Drugs/alcohol
    ○ Sexuality
    ○ Self harm and suicide
    ○ Safety
    • SSHADES
      ○ Strengths
      ○ School
      ○ Home
      ○ Activities
      ○ Drugs/substance use
      ○ Emotions/eating/depression
      ○ Sexuality
      ○ Safety
1089
Q

Differential diagnosis of adolescent jaundice?

A

Unconjugated Hyperbilirubinemia
Haemolytic anaemias, Gilberts, Crigler-Najjar

Conjugated Hyperbilirubinaemia
Viral: HAV HBV HCV HDV HEV EBV CMV HSV

Metabolic: Wilsons, Alpha-1-antitrypsin deficiency, Cystic fibrosis

Biliary Tract: stones, cholecystitis, choledochal cyst, sclerosing cholangitis

Autoimmune Liver disease: type 1 (anti-SM Ab) type 2 (anti-LKM Ab)

Hepatotoxins: Drugs eg COCP, anti-TB, paracetamol, alcohol, organophosphates

Vascular: Budd-chiari, Veno-occlusive disease

1090
Q

What prevention options can you offer to adolescents to reduce risk of disease?

A
  1. Vaccination
    * HAV
    * HBV
    * HPV
    * M Pox
    * Meningitis B Neisseria meningitidis and
    Neisseria gonorrhoeae are closely genetically
    related with between 80 to 90% sequence
    homology.
  2. PrEP and PEPSE
  3. Condoms
  4. Doxycycline PEP
1091
Q

Contraindications for ivermectin?

A

Heavy Loa Loa infection
Children <5 years old or below height 90cm
Pregnancy

1092
Q

What is the key strategy for disease control for onchocerciasis?

A

Annual community directed mass treatment with ivermectin yearly in at risk communities

1093
Q

Onchocerciasis
- Causative organism
- Vector
- Epi
- Incubation
- Pathogenesis
- Clinical features

A

Onchocerca volvulus = a filarial nematode

Vector: simulium species of blackflies

EPI: Sub-Saharan Africa by fast flowing rivers, focal disease in Yemen and Latin America

Incubation: 1-2 years, up to 4 years

Pathogenesis:
- Adult females live in subcutaneous tissue + shed living microfilariae
- Filariae migrate through skin -> often into eyes
- Most pathology caused by reactions to dead and dying microfilariae, and to endosymbiontic Wolbachia

Clinical TRIAD
- Eyes
- Sub-cut nodules
- Itchy dermatitis

Eyes: pear-shaped pupil, punctate sub epithelial stromal keropathy, sclerosing keratitis, iritis, uveitis, ‘flecked retina’, scarred choroidoretinal fundus, optic nerve atrophy

Sub-cut nodules: mainly over bony prominences

Skin: dermatitis, depigmentation of skin, thickening of skin, skin atrophy, itching and deforming

1094
Q

Onchocerciasis
- Diagnosis
- Treatment

A

Diagnosis via
- Skin-snips from vicinity of subcut nodule ?microfilariae
- Skin-lamp examination of eyes - microfilariae in anterior chamber? in cornea?
- Nodulectomy of subcut nodules for adults (not normally done)
Serology unhelpful

TREATMENT
Ivermectin -> single dose 150microg/kg PO -> repeat at 3-6 mph regular intervals eg 6 months, 1 year

Works by killing microfilariae but has little impact on adult worms apart from reducing embryogenesis -> has to be given repeatedly throughout adult life cycle of worm 10-14 years

Doxycycline -> 100-200mg/d 4-6 weeks

Works by killing wolbachia endosymbionts, permanently sterilises adult worms, blocks embryogenesis, kills ~60% adult worms

1095
Q

What are the causes of eosinophilia?

A

Infections -> ?helminths
- HIV
- Fungal eg coccidioidomycosis
paracoccidioidomycosis

Convalescence from any infection

Allergies

Drug induced

Connective tissue disease

Idiopathic hypereosinophilic syndromes

Leukaemias and lymphomas

Paraneoplatis

1096
Q

What is the common name for strongyloides?

A

Threadworm

1097
Q

How is strongyloides transmitted?

A

Direct invasion through skin
Autoinfection through intestinal mucosa or perianal skin
Faecal-oral transmission

1098
Q

Life cycle of strongyloides?

A

Invasion
Larvae migrate to lungs via bloodstream -> exit blood vessels in pulmonary alveoli -> enter airways -> coughed up and swallowed -> reach small intestine -> complete life cycle

Then
- Asexual reproduction of eggs (parthenogenesis)
- hatching of non-infectious rhabditiform larvae that are excreted in the stool
OR
larvae moult into infective filariform larvae -> penetrate gut wall or perianal skin to enter circulation

Outside body
- larvae mature into free-living male and female adult worms -> reproduce sexually or transform into infectious filariform larvae ready for re-infection

1099
Q

Symptoms of strongyloides?

A

GI
Swiftly moving cutaneous eruptions eg larva current
Asymptomatic

If immunocompromised eg steroids/chemo/malignancies etc:
- Dissemination
- Hyperinfection syndrome

1100
Q

How do you make a diagnosis of strongyloides?
Treatment?

A

Detect larvae in stool
(false negatives common)
Serology but false negatives also common and only positive 4-12 weeks after infection

TREATMENT
Ivermectin STAT dose
If not then Albendazole BD for 3 days

1101
Q

Where is Lassa found?

Animal reservoir?

Transmission?

Season?

Incubation?

Clinical presentation?

A

Where?
- Nigeria
- Sierra leone
- Liberia
- Guinea
- Benin
- Togo
- Mali
- Cote d’Ivoire
- Burkina Faso

Animal reservoir: mastomys natalensis (not rat or mouse, its a rodent)

Direct contact with infected rodent excreta, tissues, or blood or via inhalation of aerosolized virus

Poor sanitation and housing conditions correlate with greatest risk of infection
Peridomestic rodents are also part of diet in certain populations

Season: Nov and April (dry season) -> may reflect a higher prevalence of mastomys inside houses

Incubation: 3-21 days

Majority asymptomatic - mild
Severe disease: 5-10%
CSF amongst hospitalised cases

1102
Q

Cause of anthrax

A

Bacillus anthracis
Gram positive rod
Forms spores under certain environmental conditions

1103
Q

Cause of anthrax

A

Bacillus anthracis
Gram positive rod
Forms spores under certain environmental conditions

1104
Q

How is anthrax transmitted?

A

Exposure to infected animals or animal products
Also, bioterrorism risk - i.e. exposure to spores

1105
Q

How is anthrax transmitted?

A

Exposure to infected animals or animal products
Also, bioterrorism risk - i.e. exposure to spores

1106
Q

Clinical presentation of anthrax

A

95% of cases are cutaneous
Begin with painless papule, which develops into central ulcer and bursts - echar with black necrotic centre develops, which can be surrounded by small vesicles
The fever may not develop, but there may be fatigue, malaise, headache

Gastrointestinal and inhalational anthrax are also possible

Hx:
Recent exposure to infected animals or contaminated animal products
If bioterrorism risk - ?break in skin and systemic symptoms

1107
Q

Clinical presentation of anthrax

A

95% of cases are cutaneous
Begin with painless papule, which develops into central ulcer and bursts - echar with black necrotic centre develops, which can be surrounded by small vesicles
The fever may not develop, but there may be fatigue, malaise, headache

Gastrointestinal and inhalational anthrax are also possible

Hx:
Recent exposure to infected animals or contaminated animal products
If bioterrorism risk - ?break in skin and systemic symptoms

1108
Q

Differentials for cutaneous anthrax

A

Bacterial ecthyma
Rickettsial diseases
Rat bite fevers
Necrotic arachnidism
Ulceroglandular tularaemia
Bubonic plague

1109
Q

Diagnostic Tests for anthrax

A

Gram stain and culture (blood, skin lesion aspirate, pleural fluid)
Paired serological testing for antibodies to protective Ag
Full thickness punch biopsy from vesicle and eschar (fix in formalin and sent for nuclei acid amplification and immunohistochemistry)

1110
Q

Diagnostic Tests for anthrax

A

Gram stain and culture (blood, skin lesion aspirate, pleural fluid)
Paired serological testing for antibodies to protective Ag
Full thickness punch biopsy from vesicle and eschar (fix in formalin and sent for nuclei acid amplification and immunohistochemistry)

1111
Q

Treatment of anthrax

A

Treat empirically whilst awaiting results with a quinolone or doxycycline.
If disseminated, then multidrug therapy with CNS penetration.
If cutaneous than treat for 7 to 10 days.
If inhalational or risk of bioterrorism, then treat for 60 days with oral antibiotics

1112
Q

Causes of acute fever and jaundice

A

Malaria
Typhoid
Leptospirosis
Ascending cholangitis

Advanced HIV
Gram negative sepsis

1113
Q

Symptoms of pneumocystis jiroveci pneumonia

A

Sub acute onset malaise, non-productive cough, progressive exertional dyspnoea
+/- fever

O/E: desaturation on exertion
Chest may be clear on auscultation

Complications:
- resp failure
-pneumothorax

Graded as mild, moderate, severe

1114
Q

Symptoms of pneumocystis jiroveci pneumonia

A

Sub acute onset malaise, non-productive cough, progressive exertional dyspnoea
+/- fever

O/E: desaturation on exertion
Chest may be clear on auscultation

Complications:
- resp failure
-pneumothorax

Graded as mild, moderate, severe

1115
Q

Diagnostic tests for PCP

A

BAL specimens - immunofluorescence, Grocott methenamine silver staining
(Induced sputum has low sensitivity)
Chest x-ray is normal in 40%, and if positive has non-specific findings. You may see ground glass opacities

1116
Q

Diagnostic tests for PCP

A

BAL specimens - immunofluorescence, Grocott methenamine silver staining
(Induced sputum has low sensitivity)
Chest x-ray is normal in 40%, and if positive has non-specific findings. You may see ground glass opacities

1117
Q

Treatment of PCP

A

Co-trimoxazole, IV if severe, 21 days if HIV positive, 14 days if other immunosuppressive ponds
Corticosteroids, if moderate or severe (wean over 20 days)

ART asap

SEs co-trimox: cytopenias, rash, hepatitis

Second line: primaquine + clindamycin or IV pentamidine (lots of SEs)
Mild to moderate: dapsone- trimethoprim or atovaquone

Secondary prophylaxis: co-trimoxazole until CD>200 for at least 3 months (or dapsone or atovaquone)

1118
Q

Treatment of PCP

A

Co-trimoxazole, IV if severe, 21 days if HIV positive, 14 days if other immunosuppressive ponds
Corticosteroids, if moderate or severe (wean over 20 days)

ART asap

SEs co-trimox: cytopenias, rash, hepatitis

Second line: primaquine + clindamycin or IV pentamidine (lots of SEs)
Mild to moderate: dapsone- trimethoprim or atovaquone

Secondary prophylaxis: co-trimoxazole until CD>200 for at least 3 months (or dapsone or atovaquone)

1119
Q

What are the 4 key concepts within the guidelines for malaria prevention in travellers from the UK 2022?

A
  • Awareness of risk
  • Bite prevention
  • Chemoprophylaxis
  • Diagnose promptly and treat without delay
1120
Q

What are the three key drugs used for malaria chemoprophylaxis and at what stage of the life cycle do they work?

A

Atovaquone-proguanil: causal prophylaxis -> kills liver schizonts (ie pre-erythrocytic cycle)

Doxycycline
Mefloquine
Both suppressive prophylaxis -> kills RBC stage

1121
Q

What is malarone?
What regimen is it given in?
Considerations including breast feeding and pregnancy?
Side effects?

A

Atovaquone/proguanil

1 day before, OD, 7 after

Considerations:
DDI
Renal dose adjustment (eGFR<30 avoid)
Take with fatty meal or milk
Cannot crush
2nd choice pregnancy or BF

SEs:
Neuro
GI
Hair loss

CI: severe liver disease

1122
Q

Mefloquine:
What regimen is it given in?
Considerations including breast feeding and pregnancy?
Side effects?

A

250mg weekly, start 2-3 weeks before travel (to assess tolerability), continue 4 weeks after

Considerations:
DDI
Crushable
NOT for psych hx, epilepsy, diving, flying, military, cardiac, CYP3A4 eg warfarin, DOACs etc

Can use in pregnancy (unlicensed), simple paediatric administration

SEs:
Dreams
Insomnia
Anxiety
Depression 2-5x more likely
Increased gender differences -> 2x female

NB: SEs/symptoms can be seen after only SINGLE dose

CIs:
- allergy to it, ingredients, quinine, quinidine
- PSYCH Hx
- NOT IF FIRST DEGREE RELATIVES eg with epilepsy/serious MH disorders
- NOT FOR EPILEPTICS
- AVOID if serious cardiac arrhythmias

Useful in:
Babies/Pregnancy/BF
Those who can tolerate it
Renal failure
Lower income

Not useful in:
Any psychiatric/psychological Hx
Long term >1 year
Eye issues
Anticoagulants
Arrhythmias
Adventure travel – diving, flying
High stress

1123
Q

Doxycycline
What regimen is it given in?
Considerations including breast feeding and pregnancy?
Side effects?

A

Start one day before travel, take OD, continue 4 weeks after

Considerations:
* Drug-drug interactions (DDI)
* Does not reduce efficacy of OCP
* Metabolism increased by carbamazepine and
phenytoin – can double dose
* Possibly reduced efficacy oral typhoid vaccine
Can use in severe renal failure
Can use moderate liver disease (mixed UK vs US guidance)

CI: pregnancy up to 15 weeks, no children, UK – no breastfeeding (unless essential), USA – breastfeeding ok, epilepsy, porphyria

SEs:
* Oesophagitis/Gastritis
* Tooth bone effects after 15/40, teratogen
* Photosensitivity
* No antacids (make it less effective)
* Thrush
* recommended to take with food or milk

Not safe
* In epilepsy if on carbamazepine/phenytoin (can dose adjust)
* Porphyria
* Sunbather/gastritis/children/long trips/thrush

1124
Q

What antimalarials can be used for breast feeding?

A
  • Mefloquine -> yes
  • Ato/Prog -> probably but lack of data
  • Doxy -> UK says no, USA yes
1125
Q

What antimalarials can be used during conception?

A

Consider an interval before conception of:

  • Mefloquine – 3 months
  • Doxycycline – 1 week
  • Atovaquone/proguanil – 2 weeks

BUT conception on any antimalarials is not an indication for termination
Most advice based on lack of data

1126
Q

Advice to give a patient on anticoagulation if they going to malaria zone and what medications would you recommend if they are on warfarin vs DOAC?

A
  • Inform anticoag clinic 2-3 weeks before travel
  • INR before starting and one week after + intervals during travel

Warfarin
* Mefloquine-OK
* Doxycycline- may increase INR
* Atov/Prog- Not known, prog increase INR

DOAC
* Mefloquine inhibits CYP3A4 and p-glycoprotein, could increase risk bleeding
* Atovaquone minor increase CYP3A4 NOT p-glycoprotein

1127
Q

What antimalarials can epileptics take?

A
  • doxycycline YES
  • atovaquone/proguanil YES
  • chloroquine NO
  • Mefloquine NO
1128
Q

What antimalarials can you give to children?

A
  • Risk of rapid deterioration
  • Mefloquine easiest- 5kg and above, weekly
  • Atov/Proguanil- 5Kg and above with milk/yoghurt, daily
  • Doxycycline >12 years
1129
Q

What antimalarials can be given to patients with renal disease?

A
  • If eGFR<30 avoid malarone
    Can use mefloquine and doxy in severe renal impairment
1130
Q

What antimalarials can be given to patients with liver disease?

A
  • All metabolized in the liver

Severe
* Doxy Yes
* Malarone unknown
* Mefloquine No

Moderate and Mild
Can use all 3

1131
Q

What are antimalarials that are anti-causal stages vs suppressive stages?

A

Causal:
- Malarone
- Tafenaquine
- primaquine

Suppressive
- Doxy
- Mefloquine
- Chloroquine

1132
Q

Who should standby emergency self-treatment be given to for malaria?

A

Those taking chemoprophylaxis and visiting remote areas where they are unlikely to be within 24 hours of medical attention
1. Artemether + lumefantrine (4 tablets, 5 further doses of 4 tablets at 8,24,36,28,60 hours)
2. Atovaquone + proguanil (4 tablets OD 3 days)

3rd line or first line for pregnant women in first trimester (otherwise treat with regimen 1): quinine (8hrly 5-7 days) + clindamycin (8hrly, 5 days)

1133
Q

A 45 year old photographer is going on a work trip to New England in May for 4 weeks. He leaves in 3 weeks. He had a splenectomy 2 years ago after RTA in SA. He is on no additional medication, has had no advice, vaccinations or follow up in UK.
* Job for Lonely Plan

What advice would you give?

A

Additional vaccinations
* Haemophilus influenza type b
* Meningococcal ACWY
* Meningococcal B vaccine
* Influenza
* Pneumococcal

Prophylactic/standby Abx
* Pen V
* Consider standby Abx

Other infection risks
* Malaria
* Babaesiosis

Tuck trousers into socks
Permethrin spray
Tick check and removal

1134
Q

Presentation of strongyloidiasis?

A

Resp: Loeffler’s syndrome -> dry cough, interstitial infiltrates, eosinophilia

Skin: larva cures

GI: abdominal pain, indigestion, heartburn, diarrhoea, bloating

Asymptomatic

if immunocompromised:
- strongyloides hyperinfestation syndrome
- severe diarrhoea/paralytic ileum, necrotising pneumonia, gram negative sepsis +/- meningitis

1135
Q

Causes of old world/Non-American cutaneous leishmaniasis?

A

Middle East, North Africa, Asia
L major, L tropica, L infantum, L donovani

1136
Q

Causes of New world/American cutaneous leish?

A

Central and South America
L mexicana, L braziliensis, L amazonensis

1137
Q

What is cutaneous larva migrans caused by?
How should you treat it?

A

Hookworm
Ancylostoma duodenale and Necator americanus.

Tx: Ivermectin STAT or albendazole
Children -> topical thiabendazole (ivermectin)

1138
Q

What percentage of patients with latent TB go on to have active TB?

A

10%
- Pulmonary or Extrapulmonary

1139
Q

What are the two biggest risk factors for TB and in what way do they increase risk?

A

Diabetes -> increases progression from latent to active TB

HIV -> increases probability of early progression + increases probability of TB reactivation

1140
Q

What does Xpert MTB/RIF mean?

A

Tells you if TB is present
Tells you if resistance conferring mutation to rifampicin is present

1141
Q

What is the difference between auramine/fluorescence microscopy for TB vs traditional Ziel-Nielson?

A

LED fluorescence increases sensitivity from 50 to 60%

1142
Q

What are the standardised treatment regimens for TB?

A

2HRZE/4HR
(2 months all, 6 months total of iso/rif)

OR

2HRZE/4(HR)3
(2 months all, but during next 4 months give “continuation therapy” of iso/rif x3 per week rather than daily)

If patients have sputum smear status <2 (milder disease) + lack of cavitation on CXR (~50%) then 4 months treatment is non-inferior to 6 months

1143
Q

A patient has military TB and presents with seizures and a stiff neck. You suspect TBM. How would you define this patients TB?

A

Pulmonary TB

Hierarchy = PTB with EP manifestations is classified as PTB

1144
Q

What is the difference between pulmonary TB and extra-pulmonary TB?

A

PTB
- Involves lung parenchyma
- Includes miliary disease

EPTB
- Involves any other system
- Not involving lung parenchyma
- Includes:
* Hilar lymphadenopathy
* Pleural

THINK:
1. Hierarchy = PTB with EP manifestations is classified as PTB
2. For EPTB think HIV
3. Paradoxical reactions
4. Steroids

1145
Q

How long does it take for TB treatment to work?

A
  • Median time to culture conversion (eg positive to negative): 37 days
  • 90% achieved culture conversion by day 60 (Fitzwater S et al, Clin Inf Dis 2010)
  • Effective treatment reduces cough frequency
1146
Q

What is the association between onchocerciasis and epilepsy?

A
  • Associated with oncho in population based studies in Africa
    ○ Onset usually 3-18 years
    ○ Risk increases with microfilariae load
    ○ Prevalence can be high eg 18% in south Sudan
    ○ Risk reduced by biannual ivermectin and vector control
  • Historically
    ○ Dwarfism (nakalanga) with associated epilepsy reported from oncho-endemic area of Uganda/DRC historically - ?hypopituitarism
    ○ ‘nodding disease’ strange form of epilepsy seen in children in oncho-endemic areas of Uganda/South Sudan/Tanzania.
    ○ Involuntary nodding movements precipitated by food or cold.
    Aetiology uncertain - ? Cross-reactivity with developing neurones.
1147
Q

What vector is responsible for 95% of onchocerciasis transmission?

A

Simulium damnosum complex

1148
Q

Describe the vector life cycle of Simulium blackflies?

A
  • Female lays eggs under water
  • Eggs last 3 days
  • Usually 7 larval instars, develop over 7-12 days
  • Have mechanism on top to filter out water
  • Pupae - 3-5 days -> Pupate under water
  • Come out of water and fly immediately
  • Males and females mate immediately, females store sperm, often have more matings in life than need to
  • Adult females <30 days (between blood meals 3-4 days)
1149
Q

What are the skin manifestations of onchocerciasis?

A

Skin acute:
Massive skin infestations by microfilaria
Live mf cause minor symptoms
Dying mf create inflammatory reactions which provoke the reactions seen in the skin

Acute phase:
Severe Itching
Secondary skin infections due to scratching
Disturbed sleep
Poor performance
NB. These stages are reversed with ivermectin treatment

Skin chronic:
Irreversible changes
Thickening of the skin (Lizard skin)
Atrophy of the skin (The skin looks old even in young adults)
Hanging groin (Rare caused by inguinal lymph nodes dragging the atrophied skin down wards creating a large skin fold)
Leopard Skin dermatitis (the most common chronic disease)
All these conditions are associated with itching which will be relived with ivermectin but skin changes are irreversible.

1150
Q

What are the neurological consequences of onchocerciasis?

A

Neurological effects
Children
- Growth retardation (Nakalanga syndrome)
○ Huge community impact
○ Skin disease, blindness, neurological effects and chronic ill health leads to a vicious cycle of poverty
- Nodding syndrome
- Increased mortality

Adults
- OAE: onchocerciasis associated epilepsy

1151
Q

What are the eye manifestations of onchocerciasis?

A

The eye
1. Conjunctiva
- Inflammation around dying microfilaria causes pruritis and pigmentation

  1. Punctate keratitis
    - Dying microfilariae in cornea create inflammatory response -> appears as white linear opacity
    - Develops as more diffuse white spot = ‘snowflake/cotton wool keratitis’
    - As microfilaria are absorbed -> cornea regains normal appearance and transparency
    - Repeated heavy infections create irreversible changes
  2. Sclerosing keratitis
    - Permanent opacities eg 9 and 3 o’clock
    - Space between called semi-lunar keratitis
    - Rest of cornea becomes opaque
    - Pupil increasingly covered -> vision progressively impaired
  3. Chronic anterior uveitis and secondary cataract
  4. Chorioretinitis
    - Inflammatory process provokes increasingly destructive chorioretinitis with loss of vision, most often beginning at periphery
  5. Optic atrophy
    - Inflammatory changes lead to atrophy
    - Retina: clumping of pigment and atrophy of optic nerve

Major causes of blindness
- Sclerosing keratitis (difficult to see back of eye if present)
- Anterior uveitis (iritis) often associated with glaucoma and secondary cataract
- Progressive chorioretinitis
- Optic atrophy

1152
Q

How are larvicides used for vector control for Onchocerciasis?

A
  1. Use larvicides for larvae populations concentrated at identifiable points (breeding sites in white-water rapids) in rivers (eg via helicopter)
  2. ‘Safe’ insecticides eg low toxicity to non-target organisms including man, eg emulsion or as particles which larvae will filter and eat (eg Temephos and Bacillus thuringiensis serotype H14)
  3. Apply larvicides to rivers above breeding sites and rotate sites to minimise insecticide resistance. Wash insecticide down river through breeding sites -> apply for 10 minutes to ensure lethal dose reaches blackflies, 30minutes needed for S.neavei)
    ○ Idea is kills multiple breeding sites as insecticide is washed down river
    ○ Depending on river may need to treat at multiple dosing points

Other principles
- Repeat dose weekly to kill every generation (S.naevi is slower growing and insecticide can often be applied monthly)
- Measure river discharge before every treatment to ensure dilutional effect is correct
Measure success of vector control by reduction of biting flies

1153
Q

On what crustacean might you find larvae of Simulium blackflies?

A

Crabs

1154
Q

What world wide programmes exist for onchocerciasis control/elimination?
Describe community engagement within this?

A

WHO roadmap: elimination of onchocerciasis by 2030 in 12 countries. National elimination programmes are long-term programmes and based on multiple ivermectin treatments each year.

WHO’s Onchocerciasis Elimination Program for the Americas (OEPA).

Onchocerciasis control programme (OCP) = 11 countries in West Africa.

African Programme for Onchocerciasis Control (APOC) = 19 countries.
- Pioneered community directed treatment so don’t need health worker coordination etc

African programmes now nationally owned under WHO AFRO ESPEN.

Onchocerciasis Control Programme in West Africa major public success (1974-2002)
Based on vector control (larviciding initially, later on mass drug treatment with ivermectin)

Community directed treatment programmes:
1. Choose a distributor for training
2. Do a census to calculate Mectizan requirements
3. Organise the collection of Mectizan from a Health Centre or other distribution point
4. Organise a distribution method, house to house, fixed point in village etc
5. Help the distributor calculate the dose and distribute the Mectizan
6. Note the treatment statistics and report to the health authorities
7. Participate in community supervision
After 5 years communities should be ready to continue treatment alone with minimal help and supervision from the P.H.C. services
Equally a mechanism is in place for mass distribution of other medicines

1155
Q

Describe the paradigm shift in onchocerciasis control/elimination strategies?

A

FROM: CONTROL (Elimination as a Public Health Problem)
* Onchocerciasis control (elimination as a public health problem)
* Reduction of blindness and skin problems to insignificant levels
* Largely achieved except in conflict countries (An APOC success)
* However there is a constant risk of recrudescence
* Some treatment and surveillance will be needed for decades

TO: ELIMINATION (Elimination of transmission)
* Elimination is not “business as usual”
* The disease must be fully mapped to identify where
transmission is ongoing
* Treatment strategies will need to be analysed to interrupt
transmission as soon as possible

1156
Q

Why are transmission zones limited in their use for onchocerciasis control/epi?

A
  • Simulium are very mobile and with the winds can travel 400 Km so there is always a risk of reinvasion
  • Transmission can occur when treatment is not carried out “across the border” (national or provincial) with local vectors
  • may cross state or national borders and may not include whole health districts
1157
Q

Pros and cons of twice yearly treatment with ivermectin for onchocerciasis?

A

Pros
Reduces transmission particularly in early rounds (Skin microfilaria kept at zero)
May speed up the achievement
of a “closed system”
Has more effect on adult worms
May increase overall “annual” coverage
Reduces treatment time to 6 - 7 years, therefore easier to proclaim an endpoint and success in elimination

Cons
Change in CDTI philosophy
Distribution times more controlled at central level
More control from central level
No reduction in transmission after several years of treatment
Extra costs
- Retraining
- Logistics in country
More ivermectin needed

1158
Q

What is the criteria for onchocerciasis elimination regarding morbidity and transmission?

A

Morbidity
* The absence of microfilariae (mf) in the
anterior segment of the eye no longer
included

Transmission
* Presence of L3 in flies <0.05% (0.1% in parous
flies)
* ATP lower than 20 L3 per season
* Absence of detectable infection in children
<10 yrs and antibody prevalence of <0.1%
* No new infections in recent migrants

1159
Q

What is the parasitic life cycle of onchocerciasis?

A
  1. Infected Blackfly bites human
    • During the bite, fly deposits infective L3 larvae of Onchocerca volvulus into skin
  2. Larval Development in Human Host
    • L3 larvae migrate into subcutaneous tissue + grow and mature
    • Over 6–12 months, larvae develop into adult worms. Female worms can grow up to 50 cm in length, while males are smaller, reaching around 4–5 cm.
    • Adult worms form fibrous nodules or lumps, known as onchocercomas, typically around the bony areas of the skin (e.g., near the joints or skull).
  3. Production of Microfilariae
    • Inside the nodules, adult worms live for 10–15 years and reproduce continuously. Each female can release thousands of microfilariae (larvae) throughout its lifespan.
    • These microfilariae measure about 200–300 micrometers and migrate from the nodules to various parts of the body, particularly to the skin and eyes.
    • Microfilariae cause intense itching and inflammation as they travel through the skin, and can invade the eye
  4. Re-Ingestion by Blackfly
    • When an infected human is bitten by another blackfly, the fly ingests the microfilariae present in the blood or skin of the host.
    • Once inside the blackfly’s gut, microfilariae migrate to the thoracic muscles of the fly.
  5. Development of Microfilariae in the Blackfly
    • Inside the blackfly, the microfilariae undergo two molts over a period of 7–14 days, developing from the ingested microfilariae into the infective L3 larvae.
    • Once the larvae reach the L3 stage, they migrate from the thoracic muscles to the blackfly’s proboscis (mouthparts), ready to be transmitted to a new human host.
  6. Cycle Repeats with New Human Host
1160
Q

Onchocerciasis
- Cause?
- Vector?
- Transmission?
- EPI?

A

Definition
A parasitic disease caused by the filarial worm Onchocerca volvulus; primarily affects skin and eyes.

Causative Agent
Onchocerca volvulus (a parasitic worm)

Vector
Blackfly (Simulium species) = females only
Breeds near rivers and streams -> white water, fast-flowing (well oxygenated for nutrients for larval requirements)
Only bite through skin (not clothes), PAINFUL BITE, outdoors, daytime

Geographic Distribution + EPI
Mainly found in sub-Saharan Africa (96%); 4% Latin America and Yemen
21 Million WW, 123 million at risk
Blindness 270,000 (most commonly SS savanna), vision loss 1 million, skin disease 15 million

Transmission
Transmitted through repeated bites of infected blackflies; larvae enter skin and develop into adults
Major effects in skin necessary for transmission: acute dermatitis, chronic lizard skin, leopard skin, skin atrophy

1161
Q

Describe the pathophysiology of onchocerciasis

A

Adult worms create nodules in skin: <2cm diam, mobile, firm, well-defined, non-tender or painful, pelvic girdle in Africa, head and shoulders in Guatemala, Mexico and African children
Microfilariae drive disease
- Early reactions to dead microfilariae in upper dermis: eosinophil and neutrophil infiltration with macrophages – leads to itching and acute skin lesions – reversible inflammatory changes
- Neutrophils attracted by endotoxin-like molecules released from symbiotic rickettsial bacteria Wolbachia
- Later develop fibrosis, depigmentation and atrophy – leads to irreversible chronic skin lesions
- Hyperreactive form of disease Sowda: few microfilariae, high IgE levels, increased inflammatory infiltration, altered Th2 response. Intensely itchy.
- Neutrophil attraction to bacterial Wolbachia prevents eosinophil-mediated response to nematode worm: immune evasion.
- Transmitted transovarially from worm to worm (vertical transmission) = death of wolbachia (eg with tetracyclines, rifampicin and chloramphenicol) disrupts fertility and worm death

1162
Q

What is the hyper-reactive form of onchocerciasis and how does it present?

A

Sowda
few microfilariae, high IgE levels, increased inflammatory infiltration, altered Th2 response. Intensely itchy

1163
Q

What are the main symptoms and consequences of onchocerciasis?

A

Symptoms

DYING MICROFILARIAE cause DISEASE
Depend on no. of bites and severity -> related to microfilariae burden
Severe itching and acute skin lesions (rash, nodules, depigmentation)
- Early reactions to dead microfilariae in upper dermis
- Nodules found mostly on bony prominences throughout body, mostly asymptomatic
Eye lesions leading to visual impairment and, in severe cases, blindness

Consequences

Clinical
* Visual damage leading to blindness
* Itching, acute and chronic skin disease
* Increased mortality especially in <20, ?associated with epilepsy
* Clinical severity depends on length of exposure to bites and density of microfilariae in the skin. Chronic inflammatory response leads to progressive disease

Socio-economic
* Stigma + psychosocial consequences, reduced marriage consequences
* Personal and family economic impacts
* Community desertion of fertile river valleys

1164
Q

How do you diagnose and treat onchocerciasis?
What are the two key challenges of treatment?

A

DIAGNOSIS

Skin snips to detect microfilariae (normally from iliac crest)
Slit-lamp examination for eye infection.
Serologic tests in some cases
ELISA: Ov16 point of care RDT
- IgG in blood -> indicates exposure to infection

TREATMENT

Ivermectin: kills larvae, alleviates symptoms, and helps prevent transmission (given annually)
- Mass treatment every 3-6 months controls transmission as keeps MF amount low
- Mass treatment every 12 months stops blindness/clinical manifestations
○ little effect on adult worm mortality – a major drawback

Exclusion: pregnant women, lactating women within 1 week of birth, children under 5, co-infection with Loa loa

Doxycycline: depletes symbiotic bacteria (Wolbachia) within worms, weakening them (3 weeks causes sterility of adult worms, 4 weeks can kill them)

CHALLENGES
1) Loa loa co-infection (loascope can help identify patients with filarial load of 20,000 roughly as cut off -> don’t treat those with that or more), consider giving them doxy
2) Ivermectin tolerant parasites

1165
Q

What diseases does Ivermectin show effectiveness for?

A

Onchocerciasis, Wuchereria bancrofti, Ascaris, Strongyloides, head lice and scabies. Even malaria with doses every 3 weeks.

1166
Q

Who needs to be treated for onchocerciasis, ie who should we target through public health programmes?

A
  • Priority for control is communities with high endemicity (eg prevalence over 60% in adults), but need to treat all affected communities for elimination (transmission)
    • Morbidity: High prevalence → high parasite density → more pathology
    • Mass distribution (treat everybody in community regardless of infection or disease status) every 12 months for control
    • Elimination: every 3 or 6 months: reduces transmission as well as clinical benefit (very successful in Latin America)
    • Community-directed treatment is highly successful
    • Long-lived adult worms: programme must last 10 years at least (ivermectin kills mf but not adult worms)
1167
Q

What are the challenges of onchocerciasis elimination?

A

1) Capacity building (public health and lab)

2) Sustainability (length of treatment to finish task, post-treatment surveillance)

3) Conflict (population changes, infrastructure, funding)

4) Loiasis

5) New drugs *ideally want macrofilaricide but not micro, also one to combat parasite evasion

6) Data and specificity if control is being integrated into other elimination/NTD campaigns

7) Country ownership (ESPEN is facilitating national ownership but insufficient domestic funding in many countries)

8) Advocacy (need long term funding and agenda)

1168
Q

What is sparganosis caused by, where is it predominantly found, what is the life cycle/ how is it transmitted?

A

Cestode: tapeworm Spirometra

Spirometra infect animals WW, but high incidence in SE and East Asia, especially in (south and east) China (practices to do with eating frogs, snails, snakes or using their undercooked flesh to apply to skin or eyes as remedy)

Life cycle:
1) Adult worms live in the intestines of dogs and cats => release eggs into the environment via faeces
2) Eggs embryonate in freshwater, infecting crustaceans
3) develop into procercoid larva within the crustacean, then ingested alongside it by a second intermediate host, namely fish, amphibians, and reptiles.
4) Humans are accidental hosts => ingest the procercoid larvae either by drinking contaminated freshwater, or by eating or topically applying the undercooked flesh of the second intermediate host

Incubation period variable eg 1 day to 30 years -> also depends on where the spargana migrate to

1168
Q

How does sparganosis present?

What do you see on CT and MRI?

A

migratory, painful subcutaneous nodule -> like on and off lipoma
Ocular and visceral disease can occur
Cerebrospinal disease -> uncommon but well described, can affect any part of CNS, presents as seizures, headache, and cognitive decline, or focal neurological symptoms related to disease location.

CT: unilateral punctate calcification in white matter with surrounding low-density lesions

MRI: multiple nodular ring-enhancing lesions with ‘tunnel sign’ and surrounding oedema; it is more sensitive than CT

Definitive diagnosis is histological following surgical removal
Spirometra spp. procercoid larva are not distinguishable between one another morphologically and species level identification is often done by PCR or genetic sequencing
Serology is not routinely used and cross-reacts with other cestode species.

1169
Q

Options for treatment of human sparganosis?

A

Surgical removal -> most effective

Where this is not possible, high dose (≥120mg/kg) praziquantel has been used.

Observational evidence suggests that this can effect cure in some, but no randomised trial data exist. Lower doses (75mg/kg) seem to be less effective.

1170
Q

What fish are associated with ciguatera poisoning and where does it happen?
Presentation of ciguatera poisoning?

A

Amberjack
Happens in tropics and subtropics

  • D&V 3-6 hours after ingestion
  • Neurological abnormalities 3-72 hr after ingestion
    ○ Paraesthesia, pruritis
    ○ Metallic taste, painful dentition
    ○ Painful urination, blurred vision
    ○ Temp-related dysesthesias (hold-cold allodynia)
    ○ Paresis, reversible cerebellar dysfunction
    • CV: hours after ingestion
      Bradycardia, heart block, hypotension

Prognosis
- Recovery is usual
○ GI resolves within days
○ Neurological changes within weeks
Symptoms may come back/get worse if ETOH, caffeine, nuts, pork

1171
Q

How does scromboid fish poisoning present?

A

Due to poorly stored fish and build up of histamine
Presents like allergic reaction
- Rash
- Palpitations
- Tachycardia like IgE mediated immune reaction

1172
Q

How does sea urchin envenomation present? How do you manage these pts?

A

Painful
Puncture wounds
Tattoo appearance
irritating CaCO3 spines -> granulomatous inflammation
some spines loaded with venom -> localised reaction

MANAGE
Remove visible spines
hot water soak
consider tetanus vaccination, Abx
Plain X-ray
Later
- Treat infection eg bacteria, mycobacterial, fungal
surgical removal
- topical/intra-lesional steroid
- cryotherapy
- laser ablation
- conservative

1173
Q

Key questions to ask for Leishmaniasis?

A

Where are lesions?
Mucosal involvement? (need systemic tx)
Species -> need PCR as American species more aggressive
Multiple lesions?

1174
Q

Treatment options for Leishmaniasis?

A

Local:
- nothing
- intra-lesional antimony (meglumine)
- heating/freezing/surgery

Systemic treatment:
- oral miltefosine
- IV miltefosine (SSG = sodium stibogluconate) (CI: pregnancy)
- IV liposomal amphotericin B (ambisome)

1175
Q

Differentials for migratory lesions

A

Gnathostoma spinigerum
Spargonosis
Loa Loa
Strongyloides stercoralis
Strongyloides fulleborni (animal species)
Cutaneous larva migrans

Other
- Widespread eczema
- Phytophotodermatosis
- Trichinosis

1176
Q

Main causes of eosinophilic meningitis

A

Angiostrongylus cantonensis

Gnathostoma spinigerum

Baylisascaris procyonis

1177
Q

Presentation, diagnosis, treatment of gnathostoma

A

Presents:
- Acute phase: fever, urticaria, GI symptoms
- Cutenous: migratory swellings, subcut haemorrhage
- Visceral: lung, GI, CNS
- 12 years

Diagnosis
- Clinical, eosinophilia, serology (immunoblot)

Treatment
Albendazole 400mg BD 12 days

1178
Q

Differentials for rhabdomyolysis

A

Leptospirosis
Drugs
Long lies
exertion eg marathon runner

1179
Q

Differentials for traveller with leg weakness and recent diarrhoea

A

GBS eg campylobacter, shigella

Acute flaccid myelitis (enterovirus D68 infection)

Post-infectious myositis

Rhabdomyolysis eg exertion like marathon runner

HIV, syphilis

1180
Q

What are the clinical features of tetanus?

A

Initial injury or wound -> gain entry

Generalized Tetanus: Stiffness (early symptom), lockjaw (trismus), risus sardonicus (spasmodic smile), muscle rigidity, then muscle spasms -> if severe then impact ability to breathe
- Then toxin can get into autonomic nervous system -> rapid changes in HR and BP

Localized Tetanus: Muscle spasms near wound

Cephalic Tetanus: Can be localised to nerves around head and neck

Neonatal Tetanus: Generalized rigidity and spasm in newborns, often from umbilical stump infections

Modified Ablett score - to grade spasms
Grade 1: no spasms
Grade 2: Mild short-lived spasms not interfering with Respiration
Grade 3: More prolonged muscle spasm interfering with Respiration
Grade 4: Grade 3 but autonomic nervous system dysfunction

Other features
- Voice change -> muscle tension in pharynx and larynx
- Spasms -> can be mild in elderly patients
- CV instability is early
- Pyrexia -> can cause irreversible heat shock
Cardiac events common in elderly patients

1181
Q

Tetanus
What is it caused by?
How is it transmitted?
What is the pathophysiology?
What is the incubation period?

A

Causative Agent

Clostridium tetani, an anaerobic, spore-forming bacterium found in soil, dust, and animal feces
‘drumstick appearance’
- Pore forming toxin = tetanolysin and a collagenase are needed for growth and toxin production

Transmission

Entry of bacterial spores through open wounds (e.g., punctures, cuts, or burns)

Pathophysiology

C. tetani spores germinate in anaerobic conditions
Pore forming toxin = tetanolysin and a collagenase are needed for growth and toxin production
Retrograde transport into CNS along motor nerve + blocks inhibitory GABA neurotransmitters in motor neurons -> unopposed muscle contraction and spasm

Incubation Period

3-21 days (average around 8 days); shorter incubation is associated with rapid + more severe disease

1182
Q

What is the management of tetanus?

A

Management

  1. Remove toxin: Clean and debride wound to reduce bacterial load
  2. Antibiotics: Metronidazole (preferred) or penicillin
  3. Antitoxin: Human tetanus immune globulin (TIG) to neutralize circulating toxin
  4. Muscle Relaxation: Benzodiazepines (e.g., diazepam) for muscle relaxation and spasm control
  5. Supportive Care: Intensive care for respiratory support, sedation for severe cases
    • Airway
      ○ Tracheostomy is best option
    • Spasms
      ○ Benzo
      ○ Mg may help reduce requirement for muscle relaxants but not mechanical ventilation
      ○ Ventilation
    • Autonomic disturbance
      ○ Mg may help
      ○ Cooling if very hot
      ○ Only use short acting drugs
    • GI
      HAI
  6. Vaccination
  7. rehab -> patients can lose 25% of muscle during illness
1183
Q

A 25 year old male comes into the hospital with a open wound from a motorbike accident, through which he impaled his foot on a rusty fence. How should you manage his tetanus risk?

A
  • Very high risk injury
  • So regardless of vaccination status give booster and passive immunisation
1184
Q

A 61 year old female comes into GP clinic as she stepped on a dirty rock in Vietnam yesterday and is worried about her tetanus risk. She is fully vaccinated. What do you do?

A
  • Complete vaccination -> no input needed
    Even if very high risk injury if patient is fully vaccinated then no booster or passive immunisation needed
1185
Q

A 72 year old female in Indonesia breaks skin on her hand whilst sewing. Her daughter brings her to the hospital as she is worried she has not had a full course of tetanus vaccination. What do you do?

A
  • Incomplete vaccination with tetanus prone wound (but not necessarily very high risk) -> give booster
1186
Q

What is the best airway management for tetanus?

A

Tracheostomy and ventilation

To control breathing whilst muscle spasms ongoing

1187
Q

Mpox
- Causative agent?
- Epi?
- Modes of transmission?

A

Causative Agent

Mpox virus
Zoonotic Orthopoxvirus closely related to the smallpox virus (variola)
2 clades: I and II
- Clade I in predominantly Central Africa (split into a and b) -> cases increasing
- Clade II in predominantly West Africa (split into a and b)

Epi

DRC highest no. of cases, others in central and West Africa, some WW eg UK, Singapore
Clade Ia
- More equally distributed amongst men and women, general population, children especially
- Less spread outside of central Africa, but cases increasing
- Highest case fatality rate in Africa data ~4.6%
Clade Ib
- Outbreak from September 2023 onwards, started DRC, spread across East Africa and global imported cases
- Predominantly high-contact heterosexual networks
- Low case fatality <0.5%
Clade IIb
- Outbreak 2022, men >90%, MSM, men who have had recent STI, men with HIV
- Low case fatality rate <0.1%

Transmission

Animal-to-human: Contact with infected animals (e.g., rodents, primates) via bites, scratches, eating meat or handling infected materials/animal products

Person-to-person: Direct contact with skin lesions, rashes, scabs, bodily fluids, respiratory droplets, or contaminated objects (e.g., clothing, bedding -> lower risk), placental spread to fetus

Nosocomial infection can occur and via fomites

- Data strongest in clade Iib
- ?Sexual transmission 
- Transmission could be different in different clades but might also be about population representation 
- Some sexual transmission for Clade I -> not good data
1188
Q

What is the pathophysiology of Mpox?
What is the incubation period?
How does it present clinically?
How do we diagnose it?

A

Pathophysiology

Virus enters through mucous membranes, respiratory tract, or broken skin
Incubates and spreads to lymphoid tissues and blood, causing viremia and systemic symptoms
Affects skin, resulting in characteristic rash

Incubation Period

Typically 3-21 days (average 7-14 days)
Typically prodrome first then rash 1-4 days later

Clinical Features

  1. Prodrome (early symptoms): Fever, headache, muscle aches, fatigue, lymphadenopathy (swollen lymph nodes, differentiating it from smallpox)
  2. Rash: Begins 1-2 days into fever; progresses macules -> papules -> vesicles -> pustules -> scabs
    • (a few to several thousand, lesions appear and evolve simultaneously, centrifugal vs centripetal distribution)
  3. Rash typically begins on the face and spreads to other parts of the body, often including palms and soles, could be anus/vagina

Diagnosis

  1. PCR -> detect viral DNA
    • Best samples from rash eg skin lesion material/exudate, crusts, deroof lesion and take fluid from it
    • If skin lesions absent -> oropharyngeal +/- rectal swabs
      Other tests: Viral culture, serologic tests for antibodies (used less frequently in acute diagnosis)
1189
Q

What are the clinical complications of Mpox?

A

Proctitis
Genital deformity/abscesses
Pharyngitis
Lymphadenopathy
Bacterial superinfection/secondary infections
Encephalitis
Pneumonia
Higher rates of complications and mortality with HIV

1190
Q

What are the differentials for Mpox lesions?

A

Differentials

Syphilis (1o or 2o)
VZV
Disseminated cryptococcus
HSV
Disseminated gonococcal infection
Molluscum
Other causes of genital/anal ulcer diseases & proctitis eg chancroid, LGV, Behcets/IBD

1191
Q

How do you manage Mpox?
What is the prognosis?

A

Management

Assess pain (not all lesions visible o/e)
Supportive care (hydration, pain management, fever control, NSAIDs with caution)
Oral lesions - mouthwash, salt water, viscous lidocaine
Rectal - stool softners, sitz baths, topical lidocaine, avoid opioids
Antiviral therapy: Tecovirimat (Tpoxx) recommended in severe cases or for high-risk individuals
- Antiviral stockpiled for smallpox preparedness
Isolation: Infected patients should be isolated to prevent spread
Avoid scratching lesions to reduce secondary infection risk

Prognosis

  • Generally mild in most cases with full recovery within 2-4 weeks
  • Severe outcomes more likely in young children, pregnant individuals, and those with weakened immune systems
1192
Q

How can we prevent/control Mpox transmission?

A

Prevention

Vaccination: JYNNEOS=IMVAMUNE=IMVANEX (safer option) can be used in high-risk individuals or as post-exposure prophylaxis
- Live, non-replicating Vaccinia virus, 2 doses 4 weeks apart
- CI: Mpox symptoms, allergy to: egg, cipro, gent
- Can give to HIV, atopic dermatitis
- Fractional dosing (ID)

Public Health Measures: Isolation of infected individuals, contact tracing, proper use of personal protective equipment (PPE) for healthcare workers

IPC: gloves, gown, N95/FFP2 mask, eye protection, dedicated footwear, cover lesions

At home: patients manage own care, dedicated, well-ventilated room separate from others in household, cover lesions, dedicated utensils for individual eg linens, electronic devices etc

1193
Q

What is Zika caused by and what is the epi?

A

Causative Agent

Zika virus, Flavivirus
Single-stranded RNA
- Two strains: African and Asian
- Asian strain responsible for known outbreaks
- African strain -> higher transmissibility and pathogenicity
Transmitted primarily by Aedes mosquitoes (e.g., Aedes aegypti, Aedes albopictus)

Epi

2007 in Macaques + then case reported in man, similar to Dengue but different
Large outbreak 2007 Yap island (no monkeys on island, thought spread by travel)
2013 outbreak pacific islands and high cases of GBS and first evidence of perinatal infection + microcephaly
2015/2016 outbreak in Americas, growing evidence on GBS and congenital abnormalities
2016: WHO public health emergency of international concern
Few cases of Zika in UK since 2018 -> most of ours were from Americas but last year 2 from Seychelles + more from Thailand
2024: cases in North/Central/South America, some in West Africa and some East Africa, widespread also in South/SE Asia

1194
Q

How is Zika transmitted?
What is the incubation period?

A

Transmission

Mosquito bite from infected Aedes aegypti predominantly, day biting + urban environments, some from Aedes albopictus (more widespread geographically)
- Major driver in outbreaks
- Aedes can lay eggs/replicate in old tyres etc
Transplacental
Sexual transmission: Virus can persist in semen, vaginal fluids
Blood transfusion: Rare, but possible

Other: found in breastmilk but no reports of transmission, ?tears and sweat

Incubation: 3-12 days

1195
Q

What are the clinical features of zika?

A

Clinical Features

80% asymptomatic
Rash and fever most common
+/- arthralgia/arthritis, myalgia, conjunctivitis (red eyes), headache, back pain
Pregnancy complications: Linked to congenital Zika syndrome, which includes microcephaly, brain malformations, and other birth defects
- Asian strain linked to CZS, lack of data on African
- African in animal models linked to embryonic death rather than live births with neurotropism
- Risk decreases from 1st-3rd trimester (8% in first, 3.8% in third)

1196
Q

What are the complications of Zika?

A

Complications

Congenital:
Microcephaly, pointed occiput, redundant scalp skin, congenital contractures, cerebral calcifications/ventricular megaly on CT
Eye: focal retinal mottling and macular scarring, cataracts, glaucoma, optic nerve atrophy
Deafness
Seizures, tremor, hypertonia, hyperreflexia, irritability
Developmental delays, vision/hearing impairments
Neurological: Guillain-Barré syndrome (GBS) and other neurological disorders in some infected adults, post-infectious
Campylobacter risk of GBS still higher, ie GBS risk is still low
Weakness, facial palsy, inability to walk
Median onset 6 days from onset of infection to GBS
CNS involvement rare eg menin/encep

In comparison to Dengue:
- Zika: no increased in maternal s/s, Dengue: more severe disease in pregnancy, increased risk of death
- Zika: CZS, Dengue: increased risk of foetal loss

1197
Q

How do we diagnose and manage zika?

A

Diagnosis

Molecular testing: PCR on blood, urine, or saliva (best within 7 days of symptom onset, viraemia period short, 3-5 days after onset of symptoms)
- Pregnant women have longer viraemic period + longer => higher CZV risk
- Urine stays positive for long time (eg up to 3 weeks)
- Can also use semen, CSF, amniotic fluid, placental, umbilical cord tissue
Serology: Detection of Zika IgM antibodies (may cross-react with other flaviviruses)
- IgM positive first week after symptom onset + can persist for weeks, may not develop if previous dengue
- IgG appears early, may persist for years/life long, if previous flaviviruses/vaccination subsequent exposure to related flaviviruses can cause rapid and brisk rise in IgG against multiple flaviviruses

Management

Supportive (hydration, pain relief with acetaminophen, rest)
Avoid NSAIDS until dengue ruled out -> risk of bleeding complications

1198
Q

Prevention tools/advice for Zika?
What do we know about cross-protection?

A

Prevention

Mosquito control: Use insect repellent, wear long sleeves and pants, use bed nets, eliminate standing water

Travel advisories: Pregnant women advised to avoid travel to Zika-endemic areas

Safe sex practices: Use condoms or abstain from sex if partner has traveled to an area with Zika virus for several months

Pregnancy advice
If planning a pregnancy:
- Don’t travel to Zika virus endemic area ideally
- Avoid becoming pregnant for 2 months if only woman has travelled
- Avoid becoming pregnant for 3 months if both partners/ just the male partner has travelled
If already pregnant:
- Use barrier contraception if both travelled for the duration of pregnancy
If living in an endemic country:
- Mosquito avoidance

Cross-infection
Relationship is asymmetric
* Prior Zika infection – can worsen and enhance future dengue infections with Dengue 2, 3 and 4 (but not 1)
* Prior dengue infection protective for Zika
* May have implications for Zika and dengue vaccinations

1199
Q

What questions should you ask to assess the risk of a rabies bite?
What WHO categories exist to stratify risk?

A
  1. Where is the bite? Skin intact? Contamination by saliva?
  2. The circumstances of the bite or other contact
  3. Species of mammal
  4. Behaviour and appearance of the animal
    • Known rabies reservoir?
    • Animal sick or acting abnormally?
    • Bite unprovoked?
    • Not been vaccinated against rabies?
  5. Vaccination history
  6. (Laboratory test on animal’s brain)

CAT1: low risk eg drinking mild from rabid animal or animal licking closed skin
CAT2: nibbling of uncovered skin, minor scratches or abrasions without bleeding (treat as CAT3 if bat involved/if innervated areas eg head/neck/face/hands/genitals)
CAT3: single/multiple transdermal bites/scratches/contamination of mucous membranes/exposure to bats/eating raw meat of rabid animal

1200
Q

How should you manage post-exposure for rabies in patients who have
- been vaccinated
- not been vaccinated

A

VACCINATED
Cat I: wash wound, no PEP
Cat II/III: wash wound, vaccinate
1-2 visits for vaccination

UNVACCINATED
Cat I: wash wound
Cat II: vaccinate
Cat III: vaccinate and give RIG

If vaccinating ID then 2 sites and 3 vaccinates within 7 days (day 0,3,7)
If vaccinating IM then 1 site, 4 times (day 0,3,7,14-28) -> ie 3/4 visits for vaccination

1201
Q

How long can you give RIG after rabies exposure?

A

7 days after administration of first vaccine dose
Passive immunisation -> neutralise virus in wound
- Infiltrate around wound any remainder inject IM (not in buttocks)

1202
Q

A 30 year old man with HIV and a medium-low CD4 count wants to get vaccinated for rabies. What is your plan?

A

Vaccinate 3 times - 0,7,21-28 days

Check RABV neutralising Ab 2-4 weeks after vaccination to assess whether additional vaccine dose is required

1203
Q

A 72 year old woman in Thailand with RA on methotrexate gets bitten by a rabid dog. What’s your management?

A
  1. Wash wound for minimum 15 minutes with soap and water
  2. Give 5 doses of vaccine at day 0,3,7,14, 28-30
  3. Assess risk -> likely CAT II/III in this case so needs RIG within 7 days of first vaccine dose (if CAT I then no RIG)
1204
Q

What is the burden of burns ww and how should they be managed?

A

95% LMICs
· Fluid resuscitation
· Warming
· Analgesia
· Nutrition
· Infection control (including tetanus)

1205
Q

What are key surgical burdens in LMICs?
What are some of the biggest challenges/complications?

A
  • RTAs
  • Burns
  • Congenital malformations (GI>cardiac), cardiac more serious, 50% in general require immediate surgery

CHALLENGES
- lack of access
- lack of timely access
- patient factors: nutrition, anaemia, immunosuppression
- system factors: access to parenteral nutrition, safe anaesthesia, ICU, blood transfusion, hygiene, technology, Abx

1206
Q

What are the different types of FGM?

A

Type I (Clitoridectomy)
Partial or total removal of the clitoris and/or the clitoral hood.
Subtypes:
Type Ia: Removal of the clitoral hood/prepuce only
Type Ib: Removal of the clitoral glans and prepuce

Type II (Excision)
Partial or total removal of the clitoris and labia minora, with or without removal of the labia majora.
Subtypes:
Type IIa: Removal of labia minora only
Type IIb: Partial or total removal of clitoris and labia minora
Type IIc: Partial or total removal of clitoris, labia minora, and labia majora

Type III (Infibulation or pharaonic circumcision)
Narrowing of the introitus through creation of a covering seal
Layer of scar tissue formed by cutting and repositioning inner or sometimes outer labia, with or without removal of clitoral glands and/or prepuce
The vaginal opening is narrowed, leaving a small passage for urine and menstrual fluid -> ++ symptoms eg can take long time for urinate
Subtypes:
Type IIIa, removal and apposition of the labia minora
Type IIIb, removal and apposition of the labia majora

High prevalence in North-Eastern Africa: Djibouti, Eritrea, Ethiopia, Somalia, Sudan

Tx: de-infibulation = opening procedure, can alleviate some physical symptoms but cannot replace tissue that has been removed, can be done under varying anaesthetics
Re-infibulation = re-closure of women with type 3 after childbirth, illegal in UK, can happen on and off if giving birth

Type IV (Other harmful procedures)
All other harmful procedures to the female genitalia for non-medical reasons. Includes pricking, piercing, incising, scraping, and cauterizing the genital area.
May include pulling/stretching labia and clitoris or damage during sexual violence/abuse
- Stretching creates severe pain and wounds from sticks/weights to stretch labia
May be prick or small scratch -> may heal without scar but women remember it

1207
Q

What are some short term consequences of FGM?

A
  • Haemorrhage
  • Severe pain and shock
  • Urine Retention
  • Injury to adjacent tissue
  • Tetanus, HIV, Hep B/C
  • Fracture or dislocated limbs from being restrained
  • Death - severe bleeding leading to haemorrhagic
    shock
  • Neurogenic shock as a result of pain and trauma
  • Infection / septicaemia
1208
Q

What are some long term consequences of FGM?

A
  • Dysuria, recurrent uti’s, candida infections
  • Vulvadynia, clitoral pain
  • Abscesses due to infected cysts/thorns
  • Dysmenorrhoea
  • Infertility
  • Chronic PID, clitoral neuroma and chronic scar formation.
  • Sexual dysfunction and dyspareunia
  • Problems in childbirth
  • Psychological trauma
1209
Q

What are some psychological effects of FGM?

A
  • Flashbacks (maybe triggered during
    pregnancy/childbirth/suturing postpartum)
  • Sleep disturbances & nightmares
  • Panic attacks
  • Sexual dysfunction
  • Depression & anxiety
  • Touch / needle phobia
  • PTSD
1210
Q

What are some obstetric complications of FGM?

A
  • May not be identified antenatally
  • Difficulty with VE’s, applying FSE/FBS
  • Scarring & stricture of vaginal canal
  • Possible obstructed labour
  • Psychological Trauma, Flash backs
  • Limited knowledge of birth attendant
  • Increased risk of C/S, PPH, fetal
    asphyxia/anoxia & perineal trauma
  • Women with Type 3 CAN get pregnant
1211
Q

What are some treatment options for FGM?

A

Oestrial or testosterone cream
Physiotherapy - pelvic floor spasms
Dilators - to facilitate intercourse
Nerve blocks - pain therapy
Surgery De-infibulation or reconstruction

1212
Q

Describe the generalised gut coccidia life cycle

A

Oocyst ingested
Asexual reproduction or sexual reproduction in intestine, asexual leads to schizogony repeated, sexual leads to gametogony
Resistant oocytes shed in faeces
Oocyte maturation -> normally outside body

1213
Q

Name the key human coccidia

A

BIG, 2 sporocysts:
Cystoisospora
Toxoplasma

Medium, 2 sporocysts:
Cyclospora

Small, 1 sporocyst:
Cryptosporidium, Sarcocystis

1214
Q

What is the life cycle of cryptosporidium?

A

· Oocyte ingested, release sporozoites which invade gut cells
§ Small infectious dose (~100 oocytes)
· Asexual reproduction cycle in gut cells
§ Meronts with 8 units will continue in asexual cycle (type I meront)
§ Meronts with 4 units will proceed to sexual reproduction (type II meront)
§ Unlimited number of asexual cycles
· Male gametes move along gut cells to fertilise females -> microgamont produces microgametes which fertilise macrogamont of female to form zygote
Zygote develops into either thick walled or thin walled oocyst -> thin walled can cause auto infection, thin walled exits host
· Oocytes burst out of gut cells –> already infectious (allows person-person spread)
§ 80% exit host
§ 20% become thin-walled, rupture and release sporozoites –> auto-infection cycle

1215
Q

Describe the clinical features of cryptosporidium

A

o Can be asymptomatic
o Watery diarrhoea most common
o Dehydration, Weight loss, Fever, Abdo pain, Nausea/vomiting
o Usually self-limiting (1-2 weeks)
· Chronic, more severe in immunocompromised
· Can have persistent symptoms
· Can shed oocytes in stool 1 month after symptoms resolve
o Respiratory involvement
· Particularly in children, immunocompetent or immunocompromised adults
· Concern for airborne transmission
o Can be severe/life-threatening

Issues regarding longevity -> chronic diarrhoea, weight loss, other non-descript symptoms years after infection

1216
Q

Transmission of cryptosporidium?

A

· Primarily water borne
§ Including swimming pools
· Some food borne transmission
§ Unpasteurised milk
· Person-person eg Common in day care
§ More common in households where index case is <5yo
· Zoonosis from domestic/farm animals
· Airborne transmission likely

1217
Q

Diagnostic tests for cryptosporidium?

Treatment and prevention?

A

Light microscopy of faecal specimens -> acid-fast, stain with ZN/Auramine/Safarin

BRIGHT RED on ZN staining

Direct FAT, immunochromatographic RDTs
PCR/genotyping

Treatment
o No proven beneficial treatment
o Supportive care

Prevention
o Challenges
Oocytes very resistant to chlorine
Removal/inactivation of oocysts in wastewater difficult
Animal reservoirs of some species

1218
Q

What are the most common causes of human Cryptosporidosis?

A

C. parvum and C. hominis

1219
Q

Essential package of care for victims of gender based violence (including children)

A

Information : SV / DV

Medical care :
HIV PEP : within 72 hours of sexual contact

Emergency contraception : within 3 to 5 days (eg coil or medication)

Hepatitis B vaccine : within 7 days, 0/7/21 days + booster at 1 year

In endemic settings/can be in west etc: Abx eg doxy post-exposure prophylaxis (obviously not doxy if <12 years old)

Psycho-social care
- Is your patient suicidal?
- Anxiety – GAD2
- Depression – PHQ9
- PTSD – PCL5
- Referrals : refuge / counselling / social support

Medicolegal
- History of events
- Documentation
- Evidence eg swabs, clothing

1220
Q

What are causes of vaginal bleeding/blood in underwear?

A
  • Anal causes: (fissures/lacerations), constipation, inflammatory bowel disease,
    worms, trauma
  • Skin conditions: vulvo-vaginitis, lichen sclerosis, haemangioma
  • Infections: urinary tract, worms, STIs including HPV, HSV
  • Labial fusion
  • Injury – straddle injury
  • Foreign body
  • Urethral prolapse
  • Oestrogen (endocrine)
    o exogenous - neonatal oestrogen withdrawal
    o endogenous - ovarian tumour, precocious puberty
  • Exogenous medications (hormones, anticoagulants)
  • Bleeding disorders
  • Sexual abuse
1221
Q

Pathological causes of genital discharge in pre-pubertal child

A
  • Anal – IBD, infection
  • Vulvovaginitis
  • Infections
    – Streptococcus /Staphylococcus
    – STI
  • Chlamydia
  • Gonorrhoea
  • Trichomonas vaginalis
    – Candida
    – Bacterial vaginosis
    – Threadworm (symptoms)
  • Foreign body
  • Poor personal hygiene
  • Trauma
  • CSA
    – Discharge is uncommon if no abuse (seen in <1 – 2% in studies of prepubertal girls selected for non-abuse)
1222
Q

How can sexual abuse present in boys?

A

Penile discharge
* Always pathogenic (unless it’s not discharge!)
o Overgrowth, Hygiene, STI, Other infection
o +/- Balanoposthitis or balanitis
o Consider if urethral or penile?
o Urethral discharge
o is assessed by milking the length of the urethra from the base to the tip
o indicates STI

1223
Q

What are the most important human cestodes?
General treatment?

A

Taenia saginata: 77 million

Hymenolepis nana: 75 million

Taenia solinum: 10-50 million

Diphyllobothrium spp: ~20 million

Echinococcus spp: ~3 million

Spirometra spp: 1 mill

Treatment
- Praziquantel for most
Niclosamide +/- albendazole for some

1223
Q

What are the two different types of cestodes and features of each?

A

Pseudophyllids
Scolex (head): Slit-like grooves (bothria)
Uterus: No branching
Convoluted uterine tubes
forming rosettes
Proglottids/eggs: Eggs released from
proglottids (proglottids
generally remain attached
to adult worm). Eggs in
stool.
Eggs: Operculated; gives rise
to ciliated larvae

Cyclophyllids
Scolex (head): Suckers and/or hooks
Uterus: Branching +/- uterine tubes
Proglottids/eggs: Proglottids containing eggs are released from
adult worms. Both eggs
and proglottids in stool.
Eggs: Not operculated; do not
give rise to ciliated larvae

1224
Q

What Cestoda causes a B12 deficiency?
What is its life cycle?

A

Diphyllobothrium latum

  1. Definitive host: Humans or fish-eating mammals
  2. Eggs passed in feces, hatch into coracidia in water
  3. 1st intermediate host: Copepods ingest coracidia, which develop into procercoid larvae
  4. 2nd intermediate host: Fresh water fish eat copepods; larvae develop into plerocercoids in fish muscle
  5. Human infection: By consuming raw/undercooked fish with plerocercoid larvae
1225
Q

Clinical features, diagnosis and treatment of diphyllobothrium latum?

A

Clinically: mild GI eg pain and diarrhoea, vit B12 deficiency

Diagnose: eggs in stool

Mx: Praziquantel + vit B12

Control by cooking or freezing fish

1226
Q

What is the smallest tapeworm
infecting humans?

A

Hymenolepis nana

1227
Q

Features and life cycle of T.saginata

A

Taenia saginata

  • Adult worm length: 4-8 metres
  • Scolex has 4 suckers, but no hooks
  • Proglottids are broader than long, 5-10 released each day; each segment can produce 80,000-100,000 eggs/day so up to 1million

Eggs: not infectious to humans but indistinguishable to T.solinum
- Only infective to cattle
- Thick striated egg shell
- Oncosphere with 6 hooks

WW -> wherever cattle raised

  1. Definitive host: Humans (intestines)
  2. Eggs excreted in feces
  3. Intermediate host: Cattle/buffalo/illamas ingest eggs
  4. Oncospheres penetrate intestinal wall and form cysticerci in muscle
  5. Human infection occurs through consumption of undercooked or raw beef containing cysticerci (ie larvae stage)
    2-3 months for adult development in small intestine, life span 10 years+
1228
Q

Clinical features, diagnosis and managemnet of T saginata?
Control methods?

A

Gravid proglottids may crawl out of anal orifice and oviposit on perianal skin -> patient feels moving or sees in underwear

Nausea and mild abdominal
pain, and anal pruritis

Typically asymptomatic or mild symptoms such as abdominal pain, nausea, or weight loss

Eggs or proglottids in Stool

Praziquantel or Niclosamide

Control:
Meat inspection of bovine muscle at abattoirs
* Cooking meat to 56oC
* Hygiene and sanitation: environmental decontamination v. difficult, eggs are very
resistant to most environmental conditions and can survive for several years on
pastures.
Eggs are disseminated widely in the environment by water, sewage effluents,
wind, flies and birds

1229
Q

Who are T.solium ova infectious to?

A

Pigs and humans

1230
Q

Lifecycle/hosts of T.solium? Lifespan of adult worm?

A
  1. Definitive host: Humans (intestines)
  2. Eggs excreted in feces
  3. Intermediate host: Pigs or humans. Ingest eggs, which hatch in intestines
  4. Oncospheres penetrate pig gut wall, migrate, and encyst in muscles as cysticerci
  5. Human infection occurs by consuming undercooked pork containing cysticerci; larvae develop into adult worms in the intestine

OR become intermediate hosts via eating contaminated food etc containing the eggs (autoinfection also possible)

Adult worm length: 2-8m
Lifespan: 25 years

1231
Q

Clinical syndromes/signs of infection with T.solium?

Diagnosis?

Management?

Control?

A

Symptoms: WHERE are cysts? HOW MANY? Symptoms?
o Muscle – expands so no or little disease symptoms
o Brain – no ability to expand = disease
o Eye – problem usually when treated due to the inflammation

Intestinal taeniasis (through ingestion of undercooked pig meat infected with larval form causing intestinal infection with adult worm): Mild symptoms, abdominal discomfort, nausea, or diarrhea

Cysticercosis (tissue infection with larval form via ingestion of eggs from environment eg human faeces): If eggs are ingested, larvae can cause cysts in organs, leading to neurological symptoms eg CNS lesions, focal lesions, convulsions, intracranial hypertension (neurocysticercosis)

Eggs or proglottids in Stool
- Imaging (CT/MRI): For cysticercosis to detect cysts in the brain or tissues
- Serology: For antibodies in cysticercosis cases

  • Praziquantel for intestinal infection
  • PQ/ Albendazole +/- steroids for neurocysticercosis
  • Surgery in severe cysticercosis cases

Control:
Meat inspection of pork meat, cooking meat, hygiene and sanitation, antihelminthic treatment of pigs (oxfendazole), Pig vaccine (TSOL 18, Cysvax®), Good sanitation and hygiene in humans

1232
Q

What Cestode infection causes hydatid cysts? What is the life cycle?

A

Echinococcus granulosus

  1. Definitive host: Dogs and other canids eg livestock
  2. Intermediate host: Humans (accidental) or livestock (e.g., sheep) ingest eggs from contaminated food or water
  3. Oncospheres hatch in intestine, penetrates, migrate to organs (e.g., liver, lungs), forming hydatid cysts
  4. Cysts enlarge over time, causing pressure on tissues.
  5. Hydatid cysts full of larvae stages -> proliferation of protoscolices from germinal cells inside cysts -> dogs scavenge and ingest cysts
  6. Eggs can be dispersed by wind, rain, herbivores and insects. Reported to remain infective for several
    years, can survive freezing conditions.

Wild cycle: ruminant and canid (e.g wallaby and dingo in Australia)

Typical transmission: dogs ingest offal, humans accidentally ingest faeces containing eggs eg from dog

Risks: associated with sheep farming/sheepdogs or communities living close to their dogs

1233
Q

Clinical features, diagnosis, management and control of echinococcus granulosus?

A
  • May be asymptomatic for years.
  • Caused by the developing larval cyst, slow growing (but variable).
  • Symptoms vary depending on the organ involved.
  • Most common site is liver (~75% of cases), but can occur anywhere in the body.
  • Abdominal pain, hepatic mass, bile duct obstruction (liver).
  • Chest pain, cough, shortness of breath (lungs).
  • Rupture of cyst (trauma, surgery) may cause severe anaphylaxis.
  • Imaging (Ultrasound/CT/MRI): Detects cysts
  • Serology: Detection of antibodies against Echinococcus

Site/size/stage dependent
* Medical: Benzimidazole (+/- Praziquantel)
* Surgical: cyst removal, PAIR (Puncture, Aspiration, Injection (of scolicide), reaspiration)

Control: carcass removal to prevent dogs scavenging, cooking dogfood, deworm dogs, controlling stray
dog populations

1234
Q

What causes alveolar hydatid disease? What is the transmission and life cycle?

A

Echinococcus multilocularis
Small, similar to E. granulosus but more invasive

  1. Definitive host: Foxes, dogs/cats/coyotes
  2. Intermediate host: small rodents or humans (accidental) ingest eggs eg from fox poo
  3. Oncospheres penetrate the intestine and form alveolar cysts in organs, mainly liver
  4. Cysts grow aggressively, similar to a tumor, invading tissues

Typical transmission: Fox and rodents. Human: accidental ingestion of faeces (fox) containing eggs
Risk often associated with hunting, and berry and mushroom picking (contaminated by fox faeces).
Infection may also establish in domestic dogs and cats

1235
Q

Echinococcus multilocularis
- Clinical manifestations
- Diagnosis
- Treatment
- Control

A

Pathogenesis similar to E.granulosis but cysts very slow growing, often asymptomatic for 5-15 years

Tumour-like cysts, mainly in liver, symptoms: weight loss, abdominal pain, general malaise, hepatic
symptoms (jaundice etc)

Larval metastases may spread to other organs (lungs, brain, spleen) to form new cysts

If untreated -> progressive + fatal
- Imaging (Ultrasound/CT/MRI): For cysts
- Serology: Detection of antibodies
At early stage if cyst is confined: Radical surgery, followed by long term benzimidazole.

Later stages very difficult.

Control: more difficult than E.granulosis as life cycle involves wild animals. Hygiene: washing of wild
berries and mushrooms. Deworming of domestic dogs and cats may help.

1236
Q

How can humans become infected with Spirometra spp. ie get sparganosis?

A
  1. By drinking contaminated water in which there is an infected cyclops
  2. By eating infected raw or undercooked flesh (fish, frogs, turtle, snakes), containing spargana (plerocercoid larvae)
  3. By use of a poultice of raw infected flesh applied to skin, conjunctiva, or
    mucosa (traditional medicine in Asia). This allows the plerocercoid larvae in the infected flesh to crawl directly into human tissues. Raw frogs and snakes used in this way, or eaten, are the most likely reservoirs for spargana

Larvae are believed to be able to survive for up to 20 years in humans

1237
Q

Life cycle of sparganosis?

A
  1. Definitive host: canids and felines eg cats/dogs/other carnivores
  2. First intermediate host copepods
  3. Second intermediate host: fish, amphibians, reptiles

Umembryonated eggs passed in faeces, embryonate in water, coracidia hatch from eggs, ingested by crustaceans, procecoid larvae body in crustaceans, ingestion by second intermediate host, development of larvae into plerocercoid larva, ingestion of second intermediate host by predator, adult develops in small intestine

Humans become accidental hosts by ingesting infected water or undercooked meat from intermediate hosts.
Larvae migrate within the human host, sometimes to subcutaneous tissues, eyes, or CNS

1238
Q

Clinical symptoms, diagnosis, management of human sparganosis?

A

Subcutaneous sparganosis: Migrating nodules, swelling, pain
Ocular sparganosis: Eye inflammation, oedema, pain
Cerebral sparganosis (rare): Neurological symptoms, seizures

Symptoms depend on site and activity of larvae: larvae can move around body
quite vigorously (“creeping tumour”) (most common form)

DIAGNOSIS:
Imaging (Ultrasound/MRI/CT): For locating larvae.
Biopsy: Identification of sparganum larvae in tissue samples.

TREATMENT: Surgical removal of larvae

CONTROL: clean water, cooking fish/amphibian/reptile meat, avoid contact with raw
amphibian flesh

1239
Q

Brownish sclerotic, multiseptate cells are visualised under the microscope from skin scrappings. What is the likely disease and how does it present?

A

Chromoblastocystis
- Caused by various pigmented fungi
- Chronic infection of skin and subcut tissue, eg hands, feet, lower legs -> painless lesions develop and spread from site of inoculation as veraccous nodules/plaques centripetally by lymphatic or cutaneous dissemination

Complications: ulcerations, chronic lymphoedema, elephantiasis, bacterial superinfection

Tx: itraconazole PO + terbinafine/flucytosine

1240
Q

How does Konzo present?

A

SPASTIC PARAPARESIS
- occurs in cassava growing areas in Africa
- Abrupt onset of permanent but non-progressive form of spastic paraparesis
children/breast feeding mothers

  • symmetrical
  • no sensory or genitourinary involvement
  • <1 week abrupt onset with no other cause
  • cassava area with other cases usually emerging at same time
1241
Q

Hanta virus

A

Haemorrhagic fever with renal syndrome

Exposure to rodent excreta

1242
Q

What examination finding is uncommon for dengue haemorrhagic fever?

A

Jaundice

1243
Q

Incubation period of leptospirosis?

A

2-30 days

1244
Q

Cardinal symptoms of Weil’s syndrome and cause?

A

Caused by leptospirosis
Cholestatic jaundice
AKI
Haemorrhagic diathesis

1245
Q

How do you recognise loa loa in tissue and as microfilariae?

A

Microfilariae have ghost sheath

In tissues will have max 1/2 adult female worms, two uteruses and be seen in skin

1246
Q

What can chronic eosinophilia cause?

A

cardiotoxic
can cause endomyocardial fibrosis

1247
Q

Visceral leishmaniasis
- CAUSE
- VECTOR
- EPI
- RISKS
- LIFE CYCLE

A

CAUSED BY
Non-American:
L.infantum
L.donovani

VECTOR
Female Phlebotomus or Lutzomyia sandflies
- 1.3-3.5mm in length
- Weak flyers
- Only females take blood
- PAINFUL bite + INDOOR biting + feeds at NIGHT
- Peridomestic species
- Zoophilic
- Rest during day in dark, humid, sheltered sites

EPI
Globally: mass underreporting but ?up to 300,000 people per year
South Asia
- Decreasing trend towards elimination -> epidemic wave pattern every 15 years
East Africa
- No decreasing trends
- Epidemic wave pattern every 6-10 years
Latin America
- In general decreasing
- Increasing cases in Paraguay
- Brazil carries majority of cases
Endemic in parts of South Asia (India, Bangladesh), East Africa (Sudan, Ethiopia), and Latin America (Brazil); occurs sporadically in other tropical and subtropical regions.

RISK FACTORS
Risk of transmission
- Sandfly density
- Human parasite reservoir
- Proximity to dog-reservoirs
- Sandfly infection rate
- Human-sandfly interaction

Risk of disease
- Proximity to VL cases
- Poor health/immune status (malnutrition, HIV, TB and other immune-suppressive conditions)

LIFE CYCLE
1) Sandfly bite injects promastigote stage of the parasite
2) Promastigotes are phagocytosed by macrophages, transforming into amastigotes
3) Amastigotes multiply within macrophages, spreading to organs like the spleen, liver, and bone marrow

1248
Q

How do we make the diagnosis of visceral leishmaniasis?

A

Past hx of VL -> refer for parasitological diagnosis
No past hx -> rK39 RDT
- Positive -> treat
- Negative -> refer to national guidelines of relevant country
If in high HIV prevalence area -> all VL cases test for HIV and all HIV cases in endemic VL areas should be clinically screened for VL

GOLD STANDARD -> visualise amastigotes in Giemsa stained smears
- Spleen 93-99% sensitive
- BM 53-86%
- LN 54-65%

PCR
- but may identify asymptomatic people

Ag/Ab tests

1249
Q

Incubation period and clinical features of VL?

A

3-100+ weeks
Depends on patient clinical status

Clinical Signs and Symptoms
THINK: Fever>/=2 weeks + splenomegaly +/- weight loss + RFs

Case definition: fever >38 for 2 weeks + splenomegaly + rK39 positive

Other: wt loss, hepatosplenomegaly, lymphadenopathy, BM hyperplasia, anaemia, leucopenia, thrombocytopenia, hypergammaglobulinaemia, epistaxis, proteinuria, haematuria

Mortality: haemorrhages, supra-infections

Differential diagnoses: chronic malaria, typhoid, schistosomiasis, TB, hematologic malignancies

1250
Q

Differential diagnoses for visceral leish?

A

Chronic malaria, typhoid, schistosomiasis, TB, hematologic malignancies

1251
Q

Treatment options for visceral leishmaniasis?

A

Indian sub continent, L donovani:
1. Ambisome Stat high dose eg 10mg/kg or dosing to max over 3-5 days
2. Ambisome STAT + miltefosine OD 7 days OR Ambisome STAT + paromomycin OD 10 days OR Milfetosine + paromomycin OD for 10 days
3. Ambisome low dose OD for 15-20 doses
4. Miltefosine - BW dosing for 28 days

If L infantum:
1. 3-6 daily ambisome infusions up to total dose 18-21mg/kg

East Africa, L.donovani
1. Combination: pentavalent antimonials eg sodium stibogluconate (SSG) M/IV + Paromomycin for 17 days
2. Monotherapy: eg SSG or meglumine antimoniate IM/IV for 30 days
3. Ambisome 6-10 days
If L.infantum (all regions)
1. Ambisome 3-6 days up to dose of 18-21mg/kg
2. Pentavalent antimonials IM/IV over 2 days high dose or low dose 20-30 days

1252
Q

Who should NOT be given miltefosine for visceral leish?

A

Pregnant women
TERATOGENIC
need contraceptive cover for 5 months if given for 28 days and 3 months of given for 10 days

1253
Q

A patient comes back to clinic in Bangladesh two years after being treated for visceral leishmaniasis with a rash on their chin. What is this complication?

Describe its features, diagnosis, management

A

PKDL = post-kala-azar-dermal leishmaniasis

- 5-10% of treated cases, particularly in South Asia (does not self heal) and East Africa (can self heal)
- Can be years after treatment, patients otherwise well -> main issues: aesthetics and public health 
- Major reservoir of L.donovani (common in endemic areas for L.donovani) -> need to treat if transmission to be limited
- Skin rash after episode of VL
- Varying morphologies: macular (symmetrical on both sides), papular (seen on chin), nodular 
- Diagnose: clinical s/s, ?endemic area, ?hx of VL (10% will lack hx), rk39 (positive test does not confirm diagnosis), GOLD STANDARD: slit skin smear but low sensitivity 
- DDx: leprosy, fungal 
- Tx: 12 weeks Miltefosine 100mg/day WHO recommended, need >7 months contraceptive cover for females of childbearing age OR Ambisome low dose over 3 weeks, in East Africa combo treatment with antimonials/paromomycin given
1254
Q

Describe some features of the two common co-infections with VL?

A

HIV
- Both mutually reinforcing -> HIV ppl susceptible to VL and VL accelerates HIV replication and progression to AIDS
- Poor treatment outcomes and higher mortality in co-infected patients regardless of drug used
- Co-infected patients frequently relapse -> may act as reservoir for drug resistance parasites
- ?’super-spreaders’
- Threat to sustained elimination efforts
- Presentation with HIV: pulmonary/intestinal/ocular, atypical disseminated, may have no fever at presentation but exaggerated inflammatory reaction once treated
- Treatment for VL/HIV: relapse common, poor cell mediated immunity
- For both East Africa and South-East Asia: Liposomal amphotericin B + Miltefosine combination therapy recommended

TB
- 20% of VL-HIV pts co-infected with TB
- High mortality
- CBNAAT testing in all VL-HIV pts
- Low drug resistance TB prevalence but high incidence of fatal IRIS
- Poor response to monotherapy
- Delay ART>3 weeks; need 6 week admission

1255
Q

What leish species are prone to mucosal involvement in leishmaniasis?

A

Sub-genus: viannia

Leishmania braziliensis
- Found in South America, especially Brazil; can lead to mucocutaneous involvement with disfiguring lesions on the nose, mouth, and throat.

Leishmania guyanensis
- Found in the Guyanas and parts of northern South America; causes primarily cutaneous leishmaniasis but can occasionally involve mucosal tissues.

1256
Q

Diagnosis and treatment options for cutaneous leish?

A

Diagnosis: MICROSCOPY +/- PCR
Microscopy: amastigotes in skin smears or biopsies
Histology
Culture: Novy-Nicolle-Mc-Neil
Molecular Tests: PCR for DNA for species identification -> ideally fresh tissue but can do on fixed
Serology: Generally not useful in cutaneous forms, as antibody levels are often low.

Treatment
Conservative
Intra-lesional pentavalent antimonial eg sodium stibogluconate
Intravenous or intramuscular pentavalent antimonial
- SEs: Malaise and QT prolongation risk
Miltefosine (teratogenic and GI effects)
- 28 days for CL
- teratogenic
- n&v
- ocular inflammation
Intravenous liposomal amphotericin
Intravenous or intramuscular pentamidine
Paromomycin
Imidazole antifungals
Cryotherapy
Thermotherapy > 50⁰C
Surgical
Photodynamic therapy
Allopurinol

1257
Q

Cutaneous leish
- causative species
- epi
- transmission

A

Causative Species

American:
L. mexicana, L. braziliensis, L. guyanensis, and L. amazonensis
Non-American:
Leishmania major, L. tropica, L.aethiopica

The species can vary by region, with L. tropica and L. major found mainly in North Africa and the Middle East

Epi

1.5 million cases per year
- In UK travellers: 60% ‘New World’, 80% of these have mucosal involvement

Transmission

Female Phlebotomus sandflies (Old World) and Lutzomyia sandlies (New World).
1. Sandfly bites
2. Parasite enters through skin + promastigotes phagocytised by macrophages
3. Transformation into amastigotes inside macrophages + subsequent multiplication of amastigotes in cells and tissues
4. Transfer via sandfly meal
5. Amastigotes transform into promastigote in midgut -> divide -> migrate to proboscis
6. Animal reservoirs (e.g., rodents, dogs) play a role in transmission in some regions

1258
Q

Clinical manifestations of cutaneous leish?

A

Exam style: well circumscribed, central ulcerated lesion with raised borders, may have multiple if tracking along lymphatics
Cannot speciate based on lesion - ie they all look different

History:
Up to 12 weeks after a sandfly bite
Enlarging or non-healing skin lesion: ulcer / nodule / keratotic plaque
Exposed sites
Lack of response to antibacterial treatments
Recent travel

Localised (LCL)
Mucosal (ML)
Mucocutaneous (MCL)
Disseminated
Nodular lymphangitis

Lupoid leishmaniasis
Leishmaniasis recidivans
Diffuse

Post-kala-azar dermal leishmaniasis

Lesions: Typically begin as small, painless papules that enlarge and ulcerate, forming raised borders and a central ulcer. Types of Lesions: Can be dry, scaly (urban CL) or moist and ulcerative (rural CL). Location: Primarily on exposed skin (e.g., face, arms, legs). Lesions may heal over months but can leave scarring.

1259
Q

Treatment for non-American CL?

A

In immunocompetent people:
Conservative
Intra-lesional pentavalent antimonial
* meglumine antimoniate
* sodium stibogluconate
Miltefosine
Intravenous or intramuscular pentavalent antimonial
* meglumine antimoniate
* sodium stibogluconate
Intravenous liposomal amphotericin
Intravenous or intramuscular pentamidine
Imidazole antifungals
Surgical

Cryotherapy
Thermotherapy > 50⁰C
Topical paromomycin
Photodynamic therapy
Allopurinol

1260
Q

Treatment for American CL?

A

Intravenous or Intramuscular sodium stibogluconate or meglumine antimoniate
20mg/kg for 21 days

Intravenous liposomal amphotericin B 3mg/kg

Miltefosine 50 mg three times daily for 28 days
Intravenous or intramuscular pentamidine

1261
Q

Main causes of corneal scarring ww and who affected?

A

Children, leading cause of blindness in LMICs

  • Vitamin A deficiency disorders (get xerosis = dryness)
  • Measles infection
  • Ophthalmia neonatorum
  • Traditional eye medicines
1262
Q

How does corneal scarring present?

A

White pupil but IRREGULAR

ie if you get BILATERAL REGULAR whitening then more likely cataracts

1263
Q

When is the critical age for Vit A deficiency in kids?

A

6 months to 6 years
<6 months maternal vit A
in between: harder to make children eat vit A containing food malnutrition, also higher risk of malabsorption eg due to diarrhoeal diseases and higher risk of measles infection
>6 years then diet normally better

1264
Q

Four key questions to ask when examining an eye?

A

Do the eyelids open and close normally?

Is the white of the eye white?

Is the cornea clear?

Is the pupil black and circular?

1265
Q

4 key causes of a non injury acute red eye and management?

A

Acute conjunctivitis (bacterial, viral, allergic)
- normal cornea, discharge, usually both eyes
- Tx: Abx ointment 8hrly

Corneal ulcer
- Unilateral, white or grey mark on cornea
Green staining with fluorescein
If deep may see hypopyon (Can put Abx under conjunctiva eg Gent (around infection)
- Tx: hourly intensive topical Abx, antiviral for HSV (5 times a day for 10 days + NO STEROIDS)

Iritis
- Irregular pupil
- Tx: dilate pupil (break adhesions between iris and lens), steroids if sure of diagnosis

Glaucoma
- hazy cornea, dilated pupil
Diamox 500 stat then 250 x 4, may use pilocarpine x4 -> need surgery/laser

1266
Q

How to manage foreign body in eye?

A

1 Lie flat
2 Local anaesthetic drops on eye
3 Light (dark room with torch on eye)
4 Loupe (magnification)
5 Lift off foreign body
(saline wash, corner of card)

rub good eye to make bad eye lacrimal

1267
Q

Examples of injury related red eyes

A

Foreign body -> anaesthesia + light + remove

Hyphaema -> watch + wait, diamox if full to lower pressure

Perforation of globe -> refer for surgery

1268
Q

Causes of decreased visual acuity?

A

Refractive error (only one that improves with pinhole)
Corneal scar
Cataract
Chronic glaucoma
Retina and optic nerve disease

1269
Q

When do you refer for a corneal graft?

A

Corneal ulceration BILATERALLY

if one eye then focus on preserving vision in that eye

1270
Q

White of eye red, hazy cornea, pupil dilated. Diagnosis?

A

Acute glaucoma

1271
Q

Cough, conjunctivitis, coryza. Diagnosis?

A

3 Cs of measles

1272
Q

Most common cause of sepsis in HIV endemic areas/HIV patients?
Signs on presentation?

A

Non typhoidal salmonella

1/3 have splenomegaly
30% of patients will have concomitant lower resp tract infection

1273
Q

What type of bacteria is orientia tsutsugamushi?

A

Gram negative
Intracellular

1274
Q

What is the vector for scrub typhus?

A

Larval trombiculid mite

1275
Q

Epi of scrub typhus

A

1 million cases per year
2 billion at risk

Tsutsugamushi triangle: south east Asia, South Asia extending up to south Russia

1276
Q

Clinical presentation scrub typhus?
Pathophysiology and complications?

A
  • Incubation period 6-21 days
  • Fever, fatigue, frontal headaches, myalgia, cough
  • 55% painless eschar
  • Delayed generalised maculopapular rash
  • Lymphadenopathy
  • Pregnancy (stillbirth, prematurity, low birth weight in >40%)
  • Mortality 1-8% (40% untreated)
  • Immunity short lived and strain specific

Pathogenesis: Widespread vasculitis and perivasculitis, accompanied by infiltration of mononuclear cells into
affected organs (can get multi organ dysfunction)
- ARDS
- Shock
- Meningoencephalitis
- Hepatitis
- Renal failure
- Invasive ventilation
- CNS dysfunction

1277
Q

Scrub typhus treatment and prevention

A

TREATMENT
Doxy
Azithromycin
Chloramphenicol

PREVENTION
Apply repellents to boots, socks, trousers, skin
Avoid sitting/lying directly on ground
For high risk people: doxycycline 200 mg weekly (or daily anti-malarial regimen) suppresses disease but may not prevent infection (symptoms may be postponed until after cessation of prophylaxis)

1278
Q

Top 5 causes of fever from South India

A

Dengue
Scrub typhus
Malaria
Leptospirosis
Typhoid

1279
Q

Gold standard for scrub typhus diagnosis vs commonly used

A

GS: Indirect immunofluorescence assay (IFA)
(get Ag of scrub typhus and bind to Ab then fluoresce)

Can also do PCR or serology

1280
Q

Ddx for long duration pyrexia of unknown origin type illness?

A

TB
Typhoid
Visceral leishmaniasis
Brucellosis
Malaria
Endocarditis eg staph, strep, enterococcus
Q fever - Coxiella burnetti

1281
Q

Type of bacteria microbiologically for brucellosis?
What are the medically important species?

A

Aerobic
Intracellular
Gram-negative coccobacilli
Non-motile
Non-spore forming
CULTURE FOR LONG EG 6/7 DAYS

Brucella suis (pigs)
Brucella abortus (associated with cattle, can cause chronic presentations with granulomatous infection)
Brucella canis (in dogs)

1282
Q

How do you contract brucellosis?

A

Zoonosis
- consumption of unpasturised milk/diary products/under cooked meat
- contact- mucous membrane/skin penetration (vet/animal care)
- aerosolization during butchery – only 10-100 bacteria required to cause infection
-Unusual: breast milk, sex, bone marrow transplant, blood transfusion

1283
Q

What is the commonest zoonosis in humans?

A

Brucellosis
>500,000 cases each year for humans

1284
Q

Epi of brucella melitensis

A

Cause of brucellosis related to camels (feel well), goats (orchitis or miscarriages), sheep

Geographical distribution:
Europe: Greece, Spain, Italy, Portugal

Latin America: Peru, Argentina, Mexico

Arabian Countries: Iraq, Kuwait,” Iran”
* Associated with dairy products
* Men and women 1:1/Children 20%-25%
* Severe clinical disease, fever of unkown
origin, hepatosplenomegally, cytopaenias
* Rate of familial infection attack: =50%

High risk: Mongolia, China

High in UK: Somalia

1285
Q

Pathogenesis of brucella

A

Inhalation

Entry via mucosal membranes

Invasion of macrophages, reproduction, circulation in reticuloendothelial system
-> lymphadenopathy
-> invasion of BM - pancytopenia
-> invasion of spleen - splenomegaly

CAT 3 pathogen

1286
Q

Incubation period, symptoms and complications of brucellosis

A

1 week to months

SYMPTOMS
- undulant fever
- ARTHRALGIA (migratory, up to 50% patients)
- headache
- malaise
- night sweats
- depression in chronic form

Complications: HEART/BONES/BALLS
Endocarditis
Sacroiliitis
Epididymo-orchitis
Osteomyelitis
Spondylodiscitis

  • Rare <1% Acute neurobrucellosis:
    Neurologic symptoms and Guillain-Barre syndrome
  • Immune thrombocytopenic purpura
  • Rare <2% Skin manifestations
  • Ocular complications
  • Pregnancy- abortion/miscarriage

Death 2% cases (mainly due to endocarditis)

1287
Q

Investigations and diagnostics for brucellosis

A

CLINICAL
- often normal
- arthralgia
- spleno/hepato megaly
- lymphadenopathy

BLOODS
FBC
- neutropenia
less often: thrombocytopenia, pancytopenia
BIOCHEM
- raised LFTs, CRP, ESR, LDH

IMAGING
Pedro Pon’s sign on lumbar x-ray (likes lumbar spine)

MICRO
CULTURE everything you can

MOLECULAR
PCR (not great)

Serum agglutination ELISA (titre >1:160 or 1:320 in endemic area) + clinical features -> ie don’t treat positive serology only
- seroconversion and evolution of titres can be used for diagnosis

Rose Bengal test (another agglutination test)
- detects IgM and IgG (high sensitivity, high spec, good screening test in acutely unwell patients, sensitivity lower if previous infection or chronic infection, not useful for B.canis)

1288
Q

What is the leading cause
of laboratory personnel
transmitted infection worldwide?

A

Brucellosis
especially Brucella melitensis

1289
Q

How do you differentiate between typhoid and brucellosis patient?

A

Typhoid -> looks very sick

Brucellosis -> looks relatively well and may report sweat smelling funny

1290
Q

WHO guideline for brucellosis treatment

A

Doxy 100mg BD for 45 days
+
IV Streptomycin 1g OD for 15 days OR IV Gent 5mg/kg OD for 7-10 days

Relapse 5%

OR

Doxy 100mg BD for 45 days
+
Rifampicin 15mg/kg/day (600-900mg) 45 days

But relapse 5%

2nd line: co-trimoxazole

Relapse: usually defined as within 6 months

1291
Q

Prevention of brucellosis?

A
  • Wear gloves, masks and gowns when working with infected animals
  • Adequately cooking meat products, and pasteurise dairy products.
  • There are live attenuated vaccines available for animals (B.abortus and B.melitensis only), but is no human vaccine.
  • In the event of a laboratory accident or biological attack involving a Brucella species, antibiotic treatment is recommended
1292
Q

Three types of theoretical TB vaccine in terms of stage of disease?

Most promising candidate?

A

Pre-exposure
Post-exposure
Therapeutic

Candidate: M72/AS01E: but gender and age differences, some promise in reducing progression to active disease in those infected

1293
Q

What is the only licensed TB vaccine and who does it protect?

A

Bacillus Calmette-Guerin BCG
Protects children from TBM and disseminated forms
Non-specific effects
Protection from pulmonary TB lowest near equator 3

1294
Q

Who are the two key target populations for new TB vaccines?

A

Adolescents and adults (especially >15yo)
Infants eg <5

1295
Q

EPI of childhood TB

A

~10% of global cases (as per 2022 rates)
= 1.25 million children 0-14 year olds in 2022

Deaths: 214,000
- In HIV negative population 76% of these were kids <5 years
- 96% in kids who did not access treatment
- 31,000 with co-existent HIV

1296
Q

What is the epi like for primary progressive disease? (hint think of a letter to describe curve)

A

U shaped
ie high in <2yrs and then low then high in >10 years
- 50% <1 yr
25% 1-2 yr
10% >10 years

Disseminated TB in children highest in <1yrs (20%)

1297
Q

What does the Xpert MTB/XDR test for?

A

TB
extensively drug resistance TB eg to first-line and second-line TB drugs, including isoniazid, fluoroquinolones, and second-line injectable drugs (like amikacin)

1298
Q

What are the different types of PCR for TB diagnosis?

A

Xpert MTB/Rif
Xpert Ultra
Xpert MTB/XDR

Xpert MTB/RIF: Detects Mycobacterium tuberculosis and resistance to rifampicin only.

Xpert Ultra: Improved sensitivity over MTB/RIF, detecting lower bacterial loads, useful in HIV co-infections and pediatric cases; still only detects rifampicin resistance.

Xpert MTB/XDR: Detects Mycobacterium tuberculosis plus resistance to rifampicin, isoniazid, fluoroquinolones, and injectable second-line drugs—crucial for identifying extensively drug-resistant (XDR) TB.

1299
Q

What can improve the diagnosis and detection of drug resistance for TB in children?

A

TWO SAMPLES ie
Concurrent use of molecular testing on respiratory specimens AND stool

If HIV +: molecular on resp samples + stool + LF-LAM

1300
Q

How should TBM, Military TB and bone TB be treated in children?

A

HRZE 2 months
HR 10 months

TOTAL: 12 months

OR 6HRZE (6 month alternative short intensive regimen for TBM treatment)

1301
Q

How should pulmonary TB, TB lymphadenitis and all other forms of TB be treated?

A

2HRZ(E) + 2HR for children 3 months - 16 years
(add in ethambutol for children with extensive disease or high prevalence of HIV/INH resistance)

Total: 4 months

2HPMZ (isoniazid, rifapentine, moxifloxacin, and pyrazinamide) + 2months HPM for adolescents 12years and over

1302
Q

What does the TANDEM consortium look at?

A

Link between TB and diabetes

1303
Q

Why might picking up pre-diabetic individuals with TB be unhelpful in terms of treating them for diabetes?

A

Inflammation from TB drives hyperglycaemia but this reverts in some people if TB effectively treated

Ideally with screening amongst TB positive people you pick up diabetic individuals only

1304
Q

Why is treating individuals with TB-diabetes problematic?

A

Higher odds of MDR-TB
Higher mortality risk

Higher risk of poor outcome

1305
Q

Outline the general management of diabetes

A
  • Aimed at reducing short-term and long-term complications
    (cardiovascular, kidney, eye disease, foot problems)
  • Lifestyle changes
    (diet, exercise, smoking, alcohol..)
  • Glucose control
  • Cardiovascular risk management
1306
Q

What is the target glucose for patients with TB and Diabetes? What are some issues with managing TB and diabetes?

A

TB (and other significant co-morbidity): <8%
(normally <7%)

Issues:
DISEASE
Active tuberculosis
Inflammation leading to: weight loss; loss of appetite; insulin resistance

DRUG
Side-effects (eg, vomiting); drug–drug interactions; weight gain during treatment

HEALTH SYSTEMS
Access and affordability of health services;
collaboration between tuberculosis and
diabetes physicians; laboratory facilities;
continuous medicaton supply

BEHAVIOUR
Variable food intake;
physical activity;
treatment compliance

1307
Q

What is the main priority of treating a patient with TB and diabetes and what should be a key consideration?

A
  • Main consideration at start of treatment -> secondary prevention
    Consider need for anti-HTN, statins, anti-plts etc
1308
Q

How can we target DM-associated TB?

A
  1. Vaccine to prevent adult TB
    1. Prevention and better management of DM
    2. Better mx of combined TB-DM
      ○ High individual mortality
      ○ Low population/incidence effect
    3. TB preventive treatment for DM/LTBI+
      ○ High population impact
1309
Q

What are some key differences between DM-TB and DM-HIV?

A

DM-TB
~500 million at risk
~3 fold higher risk of TB
More severe pulmonary disease
~2 fold higher mortality and Rif/MDR drug resistance
More TB recurrence, PTLD, CVD and deaths post-TB
Sensitivity of IGRA/TST not affected

DM-HIV
~40 million at risk
~40 fold higher risk of TB but lower with effective ART
Fewer cavities and more disseminated disease
~2 fold higher mortality (dependent on CD4 etc)
No clear association with drug resistance
Less post-TB risk if on effective ART
Decreased sensitivity of IGRA/TST (CD4 dependent)

1310
Q

Non-infectious causes of eosinophilia?

A

Allergic disorders:
* Allergic disorders
* Asthma
* Eczema
* Hayfever
* Drug hypersensitivity

Auto-immune:
* Auto-immune Churg-Strauss syndrome
* Wegener’s granulomatosis
* Polyarteritis nodosa

Haematological
* Hodgkin’s disease
* Eosinophilic leukaemia

Dermatological
* Bullous pemphigoid
* Pemphigus vulgaris

Respiratory
* Allergic bronchopulmonary aspergillosis
* Hypereosinophilic syndrome
* Asthma

Miscellaneous
* Eosinophilia-myalgia syndrome caused by L-Tryptophan
* Spanish toxic oil syndrome
* Eosinophilic gastroenteritis

1311
Q

How many flagellae do giardia trophozoites have?

A

4 pairs ie 8

1312
Q

How do you cleanse water from Giardia?

A

Boil for 10 minutes
Chlorination doesn’t work

1313
Q

Treatment of Giardia?

A

Treatment: usually metronidazole. Mepacrine and furazolidone are also effective.

1314
Q

Bancroftian filariasis vector?

A

Various mosquitos eg culex, aedes

1315
Q

Japanese encephalitis vector?

A

Culex

1316
Q

Where is Lutzomyia vector found predominantly and what disease does it transmit?

A

Leishmaniasis

Mainly new world eg South American rainforests, Brazil

1317
Q

Infectious diseases associated with shellfish / raw fish / crustaceans

A

Clonorchis sinensis
Gnathosomiasis

Hepatitis A

Vibrio parahaemolyticus

Paragonimus westermani

1318
Q

What are Heinz bodies? In what condition might you see them?

A

oxidised, denatured bits of haemoglobin found in G-6-PD deficiency for example

1319
Q

What are Howell jolly bodies?
When might you seem them on a blood film?

A

small pieces of nuclear material usually removed from erythrocytes by the spleen so might see them if patient has had a splenectomy

1320
Q

How does a splenectomy affect the platelets?

What should asplenic patients be given and why?

A

Usually results in thrombocytosis

Asplenic patients are at particular risk of severe pneumococcal infections. They should receive Pneumovax and lifelong prophylactic penicillin. Further, if possible, some splenic tissue should be preserved :-e.g. in the rectus sheath.

Asplenic patients also suffer more severe disease following infection with Plasmodium falciparum and Haemophilus influenzae.

1321
Q

Causes of nephrotic syndrome in tropical countries?

A

*Beta-heamolytic streptococci
*Lupus nephritis
*Plasmodium malariae % (especially children 2-8yrs old, may not respond to steroids)
Amyloidosis
*Schistosoma mansoni % (tx; steroids and cyclophosphamide)
* Leprosy (due to secondary amyloidosis)
Diabetes
Multiple myeloma
Filarial worms eg Bancroftian filariasis, onchocerciasis and loiasis
Renal vein thrombosis
Hepatitis B @
Minimal change glomerulonephritis

  • = numerically important

% due to immune complex deposition
@ due to membranous glomerulonephritis

1322
Q

What live vaccines can patients with HIV have?

A

MMR
Oral polio vaccine (OPV)

1323
Q

What inactivated vaccines can patients with HIV have?

A

Pertussis
Diphtheria
Parenteral polio
Monovalent whole cell typhoid
Tetanus
Cholera
Hepatitis B
Haemophilus influensae type b (Hib)

1324
Q

What vaccines should patients with HIV NOT have?

A

BCG
Yellow fever
TY21a (oral typhoid vaccine)

1325
Q

Name some infectious conditions where corticosteroids may be of benefit?

A

Severe typhoid
Hib meningitis in children
Croup
Tuberculoid leprosy
Severe pneumocystis pneumonia
Tuberculous meningitis
Tuberculous pericarditis
Tuberculous pleural effusion
Type 1 lepra reaction
Katayama fever
VZV encephalitis

1326
Q

Name some infective conditions where steroids are not thought to benefit?

A

Meningococcal disease
Gram negative septicaemia
Herpes zoster shingles
Cerebral malaria
Visceral leishmaniasis

1327
Q

Skin features of tuberculoid leprosy and how are nerves involved?

A

1 or 2 skin lesions which are well-demarcated, dry, scaly, hypopigmented, anaesthetic and hairless

ASYMMETRICAL nerve involvement

1328
Q

What happens to the platelets in clinical malaria?

A

Thrombocytopenia

1329
Q

What is the infective period for each of the following conditions?
- chickenpox
- measles
- mumps
- rubella
- bordatella pertussis
- scarlet fever

A

Chicken pox: 5 days before rash to six days after last crop

Measles: from onset of prodrome until 4 days after onset of rash

Mumps: 3 days before until 7 days after salivary swelling

Rubella: 7 days before onset of rash until 4 days after onset of rash

Bordetella pertussis (whooping cough): 1 week after exposure to 3 weeks after onset of symptoms. The period of infectivity may be shortened by antibiotics

Scarlet fever: from appearance of rash until completion of 1 day’s penicillin

1330
Q

Features of amoebic liver abscess

A

Amoebic liver abscess is the most common extra-intestinal manifestation of amoebiasis. All age groups may be affected but occurs most commonly in men aged 20-60 years

In approximately 50% of cases there is no previous history of amoebic dysentery.

The patient typically presents with fever and right upper quadrant pain of fairly acute onset. Treatment of the fever with chloroquine may modify the clinical presentation.

Neutrophilia is usual, NOT eosinophilia.

The right hepatic lobe is usually affected. Abscess formation in the left lobe is an indication for drainage.

1331
Q

Infections with incubation periods of <10 days?

A

dengue fever
yellow fever
rickettsial infections
plague
falciparum malaria - the incubation period usually quoted for falciparum is 10-14 days. Occasionally it may be as short as 8 days.

1332
Q

Infections with intermediate incubation periods (10-21 days)?

A

malaria
African trypanosomiasis
enteric fever (7-21 days)
brucellosis
hepatitis A (2-6 weeks)

1333
Q

Infections with long incubation periods?

A

brucellosis
TB
HIV
filariasis
leishmaniasis
amoebic liver abscess
hepatitis B (45-160 days)

1334
Q

What treatment is recommended for typhoid fever?

A

Cipro or 3rd gen cephalosporins (due to widespread resistance to penicillins)

In severe cases add dexamethasone

1335
Q

Three common complications of typhoid?

A

Jaundice (as a result of haemolysis, hepatitis, cholecystitis or cholangitis)

Myocarditis (3rd week)

Bowel perforation occurs in 5% of cases in the third week and has a high case fatality rate. Surgical intervention is thought to improve prognosis.

1336
Q

What should Babies born to hepatitis B antigen positive (HBeAg+ve) mothers be given?

A

Passive and active immunisation to reduce risk of vertical transmission

(additional vaccinations at birth, 4, weeks and 1 year)

1337
Q

A sudden deterioration in a previously stable hepatitis B carrier should suggest the possibility of …

A

delta virus super-infection. This scenario is especially common in intravenous drug abusers.

1338
Q

Which hepatitis virus is most implicated in HCC worldwide?

A

B

1339
Q

E.coli 0157 / H7 causes?

A

Haemorrhagic colitis

Can cause HUS

1340
Q

EBV associated cancers?

A

nasopharyngeal carcinoma

Intra ocular lymphoma

Burkitt’s lymphoma

1341
Q

What cancer is Clonorchis sinensis associated with?

A

Cholangiocarcinoma

1342
Q

Schistosoma haematobium is associated with which cancer?

A

Squamous cell of bladder

1343
Q

A positive Leishmanin test indicates?

A

Good cell mediated immunity to Leish

  • will be negative in VL but can become positive after treatment
1344
Q

Name two cell based results in visceral leish?

A

polyclonal hypergammaglobulinaemia

1345
Q

unilateral slapped cheek in a child is suggestive of what condition?

A

Haemophilus influenzae septicaemia

1346
Q

What condition gives children bilateral slapped cheek appearance?

A

Erythema infectiosum
Fifth’s disease
Caused by Parvovirus B19

Exanthem = maculopapular rash (face -> trunk -> limbs) after between 4-14 days during which time they are infectious

30% cases asymptomatic, Ab prevalence against infection in UK is high

Self-limiting arthritis in adult women can occur

1347
Q

Who is Parvovirus B19 infection dangerous for and why?

A

May cause aplastic crises in sickle cell disease and in other hereditary haemolytic anaemias

It may also be responsible for refractory transfusion dependent anaemias in immunocompromised patients

1348
Q

Is parvovirus B19 infection safe during pregnancy?

A

Known to cause hydrops fetalis (especially in 2nd trimester)

1349
Q

What are the two types of IGRA for TB?

A

QuantiFERON-TB Gold (QFT): Measures interferon-gamma released in response to Mycobacterium tuberculosis antigens. A blood sample is incubated with TB antigens, and the response indicates TB exposure. This test requires a lab with specific processing equipment.

T-SPOT.TB: Measures the number of immune cells producing interferon-gamma when exposed to TB antigens. It’s especially useful for immunocompromised patients and has a slightly different sensitivity profile.

1350
Q

How does an IFN test actually work, what are the questions to ask to interpret the results and what is the cut off?

A

Nil tube (grey):
○ Background IFN-Y

Mitogen (purple): maximally stimulates T cells to release IFN-Y
○ Positive control

TB1 and TB2 -> peptides that are specific for mycobacterium tuberculosis

Q1. is mitogen [IFN] above threshold?
– YES continue to read TB and control tube
– NO INDETERMINATE result STOP

Q2. what is TB [IFN] minus control eg nil tube [IFN]?
– < 0.35 = IGRA NEGATIVE
– ≥ 0.35 = IGRA POSITIVE

1351
Q

What does a low mitogen response in IGRA mean, ie if it is not above the threshold?

A

Could indicate an inadequate immune response (e.g., due to immunosuppression or poor sample handling) -> ie person does not generate a good IFN=Y response

In such cases, the IGRA test may return an indeterminate result because the IFN-γ response cannot be reliably interpreted, potentially masking a true TB infection status

1352
Q

Who should be treated for latent TB infection and with what?

A

Who:
- Children
- Household contacts
- Immunosuppressed

What:
- Isoniazid for 6-9 months
- Rifampicin + Isoniazid for 3 months
- Rifampicin for 4 months

1353
Q

What is a trial that suggested latent TB regimen should be shortened and to what?

A

Three Months of Rifapentine (P) and Isoniazid (H) for Latent Tuberculosis Infection

Once-weekly RifP + Iso given for 12 weeks compared to OD isoniazid for 9 months

RifP/Iso was found to be non-inferior to Iso alone and had better compliance (4 countries, >7000 people)

ANOTHER TRIAL:
1HP OD for prevention of HIV-TB -> given Daily HP for 4 weeks v daily H for 36 weeks -> same TB prevalence ie no difference ie non inferior

1354
Q

What is the TRUNCATE-TB strategy trial ?

A

Trial to see if 2 month regimen for TB was as effective as standard 6 month regimen

674 patients who were not resistance to rifampicin, treatment length could be extended if necessary

4 strategy groups were trialled but only non-inferiority assessed in 2

Two options both included (isoniazid, pyrazinamide, and ethambutol), varied based on:

  1. Bedaquilline + linezolid

initial treatment with an 8-week bedaquiline–linezolid regimen was noninferior to standard treatment for tuberculosis with respect to clinical outcomes

  1. Rifampicin + linezolid

did not meet same success rate

Both had similar level of adverse events to standard therapy

1355
Q

Infectious causes of spastic paraparesis?

A

Syphilis
HIV
TB
Neurobrucellosis
HTLV-1
Schistosomiasis

1356
Q

Non-infectious causes of spastic paraparesis?

A

Konzo
B12 deficiency
Trauma
MS
Primary lateral sclerosis
Familial spastic paraplegia
Anterior spinal thrombosis

1357
Q

HTLV-1
- EPI
- Transmission
- Diagnosis
- Treatment
- Complications

A

Human T-lymphotropic virus-1
7 types, most type A
RNA retrovirus, spread by cell-to-cell contact

  • 5-10M carriers globally. Geographical foci: Japan, Latin
    America, Caribbean, Africa, Central Australia

Transmission:
Parenteral: blood transfusion, transfusion of cells, IVDU, solid organ transplant

Mother-to child: breastfeeding, rare before and during childbirth

Sexual

PRESENTATION
- most asymptomatic (95%)
- means widespread family involvement and easily transmitted

Skin effects:
- eczema
- seborhoeic dermatitis
Rheum effects:
- sjogrens syndrome
- arthropathy
- polymyositis
Lung effects:
- Bronchiectasis
- Interstitial pneumonitis

DIAGNOSIS
Serology EIA/Western blot, Proviral PCR

TREATMENT
nothing and no vaccine

Dermatitis common in kids -> need long term Abx therapy eg co-trimox and consider mild topical steroids + follow up

COMPLICATIONS
- Most highly ONCOGENIC VIRUS in world -> T cell leukaemia/lymphoma
- HTLV-1 associated myelopathy (HAM)/Tropical spastic paraparesis (TSP)
INFECTIVE DERMATITIS especially in children

PREVENTION
- Screen blood/organs
- No breast feeding if secured alternatives are in place
- Condom use

1358
Q

HTLV-1 associated myelopathy (HAM)/Tropical spastic paraparesis (TSP)

A

PATHOPHYSIOLOGY
Abnormal activation of cytokines -> mass inflammatory effect

Chronic inflammation of white and grey matter of lower thoracic spinal cord
(infiltration of T cells– bystander effect / auto- antibodies) followed by
atrophy

FEATURES
Geographical variation
~0.25% Japan, ~3% elsewhere
* Discrepancy striking motor
symptoms > mild sensory disturbance (slow, mild, v fast progressors)
* Early signs: Lumbar pain, bladder disorders, repeated falls due to “clumsy legs”

RFs:
Living in endemic country
40-50 years old
Genetic predisposition
High HTLV-1 provirus load
High HTLV1 Ab titres

CNS effects:
- encephalopathy, leukoencephalitis, low back pain, constipation, unstable bladder

Treatment
- 1g IV methylprednisolone (3 days) for symptomatic relief but screen first for co-infections eg strongy etc
- No role for antiviral therapy or IFN-Alpha
- Symptomatic support: antispasmodics, laxatives, physio, secondary infections

1359
Q

Presentation/complications of HTLV-1 associated T cell leukaemia/lymphoma

A

Aggressive proliferation of mature
activated CD4+ CD 25+ T-cells

Long latency period - can be infected for 20-50 years before presentation
Median age 56 years

Clinical Presentation:
Lymphadenopathies, heptaosplenomegally, visceral lesions

Diagnosis: ”Flower cell” on peripheral blood film = acute ATL patient- leukemic cells with multilobulated nuclei

Complications: Paraneoplastic hypercalcaemia, opportunistic infections eg TB, strongyloidiasis

1360
Q

WHO definition of anaemia

A

Hb< 12g/dL in women
Hb<13g/dL in men

1361
Q

Biggest causes of anaemia world wide

A
  1. Dietary iron deficiency (at least 40%)
  2. Haemoglobinopathies and haemolytic anaemias
  3. CKD /Endocrine, metabolic, blood, immune disorders
1362
Q

Common causes of anaemia?

A
  • Iron/haematinic deficiencies - consider “trial of treatment”
  • Haemoglobinopathies/haemolysis
  • Chronic disease
  • Infections/infestations
  • Bleeding – take a good bleeding history, including thorough menstrual history
    • (Haematological cancers are much less common than all of the above)
      Assess for other cytopenias/features of haematological disease
1363
Q

Management of Life-threatening/severe
anaemia with haemodynamic
compromise?

A
  • Transfuse RBC/whole blood
  • Manage any bleeding
  • Once patient stable, likely to need
    additional iron supplementation
1364
Q

Management of non-severe anaemia?

A
  • Oral iron supplementation
  • Once daily/alternate day dosing
  • 40-80mg elemental iron likely
    adequate
    (NB: more you give it, less you absorb it)
  • IV iron supplementation (if available)
  • May be helpful in:
  • Surgery planned within few weeks
  • Lack of response to oral
  • Malabsorption
  • Dialysis dependent renal disease
  • Treat for 6 months (and min. 3/12 post
    normalisation of FBC)
1365
Q

When should you transfuse a patient with IDA?

A
  • Severe symptomatic anaemia
  • Transfusion has clear risks and complications
  • Transfuse the minimum amount of blood necessary to improve the clinical condition
  • Short term fix -> Always aim to correct the underlying cause
  • Always weigh up risks vs. benefits
1366
Q

What are some medical and surgical alternatives to blood transfusion?

A

Medical
* Haematinic replacement
* Anti-fibrinolytics e.g. tranexamic acid
* Erythrocyte stimulating agents e.g. erythropoietin

Surgical
* Cell salvage
* Surgical techniques to minimise blood loss

1367
Q

If DVT remains untreated, what is the risk of PE?

A

15–24%

1368
Q

What are the issues when considering treating DVT in pregnancy?

A

Anticoagulation
* Can prevent potentially fatal PE
* Increases risk of bleeding – especially complicated around time of delivery

LMWH:
* Safest but expensive

UFH:
* Can be used in pregnancy but needs some APTT monitoring and risk of Heparin-Induced
Thrombocytopenia and Thrombosis (HITT) and osteoporosis

Warfarin:
* Teratogenic in 1st trimester
* Increased risk of fetal bleeding throughout pregnancy
* Needs stopping at least 2/52 prior to delivery as crosses placenta -> unacceptable bleeding
risk for fetus during delivery

DOACs
* Not enough evidence yet to recommend
* Rivaroxaban shows fetal toxicity in animal studies, apixaban does not

1369
Q

What is a key manifestation of HTLV-1 infection in children?

A

Infective dermatitis (3-15 year olds)

  • Lesions of the scalp, ears, retroauricular areas, paranasal and perioral areas, neck, axillae, thorax,
    abdomen, groin, and other sites.
  • Lesions may be erythematous/scaly, exudative and/or crusted
  • Chronic, relapsing condition; prompt response to therapy, but recurrence upon discontinuation of
    antibiotics
  • Lymphadenopathy
  • Failure to thrive

TREATMENT
* Long-term antibiotic therapy
(trimethoprim-sulfamethoxazole);
* Mild local corticosteroids for severe cases
* Follow up

1370
Q

What is the relationship between Strongyloidiasis and HTLV-1?

A

Mortality: 15-87%

Hyperinfection risk:
- Recurrent Gram-negative sepsis
- Associated with normal eosinophils

Lots of other complications eg:
- ARDS
- Larva currens
- Nephrotic syndrome
- Small bowel obstruction

Diagnosis: direct examination, serology
Treatment: Ivermectin

1371
Q

Acronym for causes of eosinophilia?

A

CHINA
Connective tissue disease
Helminth
‘Idiopathic’
Neoplastic
Allergic

1372
Q

Describe the clinical presentation of acute lymphoblastic leukaemia related to cell counts?

A

Leukopenia
- Neutropenia
- Fever
- Infections
Thrombocytopenia
- Bruising
- Bleeding
Anaemia
- Pallor
- Fatigue
- Weakness
- SOB
Leukocytosis
- Bone pain
- Stroke
- CN palsy
- Renal failure
Extra-medullary sites
- Generalised lymphadenopathy
- Hepato-splenomegaly
B symptoms
- Weight loss
- Drenching night sweats
- Unexplained fever
- Bone pain

1372
Q

Presentation of Non-Hogkin’s lymphoma

A

Most common between age 5 and 15, almost never under 3yrs

Symptoms usually RAPID in onset (<3 months) including:
Rapidly growing mass most commonly affecting:
* Jaw, retro-orbital, LN’s, Mediastinum, abdomen – liver, LN’s Spleen, Kidney

B-symptoms
* Night sweats
* Bone pain
* Weight loss
* Unexplained fever

CNS/PNS signs
* Central nerve palsy
* Lower limb weakness
* Constipation
* Incontinence – urine/stool

Signs of bone marrow failure
* Anaemia
* Neutropenia
* leukopenia

1373
Q

Presentation of Hodgkin’s lymphoma

A

Symptoms tend to be many months (sometimes years before presenting)

Symptoms include:
Slow growing Mass
▪ Swollen LN (groin, neck, chest,armpit)
▪ Hepatosplenomegaly

B-Symptoms:
▪ Night sweats
▪ Bone pain
▪ Weight loss
▪ Unexplained fever
Recurrent infections

1374
Q

Presentation of retinoblastoma

A

Symptoms involving one eye or both include:
Leukocoria - white pupil that does not reflect the light
Strabismus - eye to become crossed because of loss of vision
Painful red eye
Decreased vision in one eye
Eye swelling or proptosis

Most common in infants and young children (almost all diagnoses before age six years)

1375
Q

Presentation of Wilm’s tumour

A

Kidney tumour

Symptoms/signs include:
Child is generally well
Abdominal mass +/- pain
Haematuria
Urinary tract infection
Hypertension

1376
Q

Presentation of brain and spinal cord tumours (especially in children)

A
  • Persistent headache
  • New onset seizures
  • Raised ICP signs/symptoms
  • Headache and vomiting on waking or early morning
  • Visual disturbances
  • Cranial nerve abnormalities,
  • Gait abnormalities eg Ataxia
  • Changes in continence
  • Deteriorating school performance or developmental milestones
  • Behaviour changes – lethargy, aggression

If fast progression -> more likely to be aggressive disease

1377
Q

Which malignancy is more rapid, non-Hodgkin’s vs Hodgkin’s?

A

Non-Hodgkin’s

1377
Q

What are therapeutic vaccines for TB?

A

aim to improve outcome of treatment of active disease
added in to standard drug treatment
tough to do as standard treatment already works well

1378
Q

Causes of B12 deficiency

A

Infection: H pylori, Giardia, Diphyllobothrium latum

Malabsorption: pernicious anaemia

Co-morbidities: gastric resection/banding, coeliac, tropical spreue

1379
Q

Features of G6PD deficiency?

A

X-linked
Female carriers have increased resistance to Malaria
Variable phenotypes: eg mild in African patients, moderate in Asian, severe in Mediterranean

Clinically:
- Neonatal jaundice: peaks 2-3 days after birth, variable severity
- Acute haemolytic: triggered by anti-oxidant drugs eg primaquine, food, infections
- Chronic non-spherocytic haemolytic anaemia

1380
Q

Primary ITP definition?

A

Immune thrombocytopenia
Acquired immune mediated disorder characterised by platelet count <100 and absence of any obvious initiating or underlying disorder

can be
- newly diagnosed
- persistent: 3-12 months
- chronic >12 months

1381
Q

Infections where ALT>AST

A

Viral hepatitis

1382
Q

Infections where ALT>AST

A

Yellow fever infection

Disseminated herpes simplex (eg in HIV immunocompromised patient)

1383
Q

Yellow fever
- Cause
- Vector
- Epi

A

CAUSE
Yellow fever RNA virus, flavivirus, 7 serotypes (vaccine protective against all)

VECTOR
Aedes aegypti (widespread across world)
Haemagogus in South American jungles

EPI
Yellow fever belt across mid of Africa and South America

1384
Q

Describe how yellow fever is transmitted and factors influencing its transmission?

A

Transmission
Mosquito bites (primarily Aedes spp.); cannot be spread person-to-person directly

  1. Sylvatic (Jungle) cycle
    • Virus transmits between mosquitoes and non-human primates
  2. Anthropogenic sylvatic cycle
    • Humans in forest get disease eg sporadic cases, can transmit back to mosquitoes
  3. Village epidemic
    • Viraemic humans go back to villages eg from forests and mosquitoes pick virus up (small outbreaks)
  4. Urban epidemic
    • Feared due to mass epidemic potential

Africa: 1-4
Only 1 and 4 in South America

1385
Q

Incubation period and clinical presentation of yellow fever?

A

INCUBATION PERIOD

Typically 3-6 days after mosquito bite, can be longer (<21 days)

DISEASE PHASES

Many asymptomatic or mild febrile illness
1. Acute phase: fever, muscle pain, headache, chills.
2. Toxic phase (0-8-7.5% progress): jaundice, organ failure, hypotension, shock, haemorrhage, liver failure 30-60% CFR
- AST>ALT *only number of infections that can do this, other one is HSV in immunocompromised patients
- Little detectable virus but immune complex deposition

RFs for mortality based on Brazil study: older (>45 yrs), neutropenia, AST>3500, hyperbilirubinaemia, high RNA levels, renal dysfunction

SYMPTOMS

Fever, chills, headache, back pain, nausea, vomiting, jaundice (yellowing of skin/eyes), and bleeding

1386
Q

What is HLH?
Potential therapies for HLH?

A

Haemophagocytic lymphohistiocytosis
- cytokine storm + macrophage engulfing of other blood cells

IVIG, anakinra (recombinant human IL-1 receptor antagonist), high-dose methylprednisolone

1387
Q

Diagnosis, treatment and prevention of yellow fever?

A

Diagnosis

Serology (IgM ELISA)
PCR testing
Virus isolation

Treatment

No specific antiviral; supportive care for dehydration, fever, and organ support if needed

Potential new therapy: Galidesivir
- Small molecular antiviral, nucleoside (adenosine) analogue
- Broad-spectrum in vitro against RNA viruses
- Survival benefit in animal models of Ebola, Marburg, Zika, YF
- Has been used in cases of YF vaccine with thymectomy

Prevention

Vaccination (highly effective, typically lifelong immunity)
Mosquito control
Protective clothing

Vaccine

Live attenuated vaccine (Yellow Fever 17D strain)
- No ability of infect vectors
- >90% protection after 10 days, >99% protection after 30 days (likely life-long protection)
- Recommended for endemic regions and travellers
Needs cold-chain

1388
Q

Microbiological characteristics of bartonella?

3 key species that infect humans?

A

Gram neg, facultative intracellular bacteria

Bartonella
- henselae
- quintana
- bacilliformis

1389
Q

Pathogenesis of bartenellosis

A

¡ Invades endothelial cells and erythrocytes
¡ Induces angiogenesis via VEGF and other growth factors
¡ Avoids immune clearance by intracellular localization
¡ Chronic infection through persistent bacteremia
¡ Subversion of immune system: Inhibition of apoptosis in host cells
¡ Bacterial virulence factors

1390
Q

Bartonella henselae
- Transmission
- Geographical distribution
- Symptoms
- At risk groups

A

Transmission

Cat scratches or bites, flea exposure

Geographic Distribution

Worldwide, especially in areas with high cat populations

Symptoms

Immunocompetent: CSD: regional lymphadenopathy, fever, fatigue, headache

Immunocompromised: bacillary angiomatosis: red vascular skin lesions, also can be in bone/LNs, can be life threatening

High-Risk Groups

Children, cat owners, immunocompromised individuals

1391
Q

What are the associated diseases of Bartonella henselae?

A

Cat scratch disease

Immunocompromised: bacillary angiomatosis

Parinaud’s oculoglandular syndrome

1392
Q

Diagnosis and treatment of bartonella henselae associated conditions?

A

DIAGNOSIS
Serology for Bartonella henselae
PCR for bacterial DNA
Biopsy for angiomatous lesions (Warthin-Starry stain)

TREATMENT

CSD
* Patients with lymphadenitis: 5-day course of azithromycin
* Patients intolerant to azithromycin: clarithromycin, rifampin, or trimethoprim-sulfamethoxazole

Bacillary Angiomatosis
* Macrolides: erythromycin or doxycycline
* Patients intolerant to erythromycin or doxycycline: azithromycin or clarithromycin
* Mild cutaneous BA can be treated for 12 weeks
* If the response is not satisfactory, the duration of therapy can be extended
* The duration of therapy has not been determined and most likely longer in cases with invasive disease

1393
Q

What Bartonella species is endemic to Andes and what condition does it cause?
What are the microbiological features?
How is it transmitted?
Who is most at risk?

A

Bartonella bacilliformis
- Flagellated, gram-negative, facultative intracellular, aerobic
- 2-3 micrometres

Carrion’s Disease (Oroya Fever and Verruga Peruana)

Vector-borne disease
Possible Lutzomyia bites (in Andes regions)
No evidence of animal host

Geographically confined
Endemic to the Andes region (Peru, Bolivia, Ecuador, Colombia)

Children in endemic countries most at risk (?lack of immunity from repeated infection)

1394
Q

Pathogenesis of Bartonella bacilliformis?

A
  • B. bacilliformis infects erythrocytes and endothelial cells
  • Physical damage and introduction of antigens into the erythrocyte membrane, producing intense erythrophagocytosis

Result: hemolytic anemia

Virulence factors (adhesin, flagellin and hemin): evade the host immune response

Acute phase: ⬆ interleukin-10 secretion by erythrocytes, decrease cytokine production → suppresses T helper cells, macrophages, and dendritic cells

¡ favors persistent B. bacilliformis infection and increase the risk of secondary infection

1395
Q

Clinical presentation of bartonella bacilliformis?

A

= Carrion’s disease
Acute: Oroya fever
Chronic: Peruvian wart

Biphasic:
acute febrile phase (Oroya Fever)
- can also be asymptomatic
- Fever + haemolytic anaemia
- Mortality 44-88% (esp without Abx therapy)
- Acute bacteraemia for 60 days (10-210)
- Complications include super-infections eg Salmonella, Toxoplasma, Histoplasma, haem, GI, CV !! Eg HF, pericardial effusion, Pul oedema, Neuro

later

eruptive/chronic phase (Peruvian wart) with red skin lesions
- weeks to months after acute phase
- classified as: Miliary, mular, diffuse
- worse in young patients eg children
- ?reservoir of disease

Risk factors for mortality in acute phase:
age >45 years, history of alcohol use, shock,
pulmonary edema, pericarditis, seizures, coma, anasarca, petechiae,
and altered mental status

1396
Q

Diagnosis of bartonella bacilliformis?

A

Acute:
Blood smears: low sensitivity
Blood culture: >70% culture positive, takes long time (>14 days)
PCR
Immunological tests: sens + spec
- Can reflect persistent humoral immune response
- IFA (indirect fluorescence Ab test)
Immunoblot-based protocol
- Potential for new ELISA

Chronic:
Clinical +/- skin biopsy
Culture: 13-47% positivity (can have asymptomatic or transient bacteraemia)

1397
Q

Treatment of bartonella bacilliformis?

A

Acute:
*Ciprofloxacin or chloramphenicol
Ceftriaxone (severe cases in children/pregnant women)
Co-trimoxazole (uncomplicated cases)

Chronic:
Azithromycin

AMR: rising cipro resistance

1398
Q

What conditions does Bartonella Quintana cause and how do they present?

A

Trench fever
Bacillary angiomatosis
Endocarditis

Incubation 5-20 days

Trench Fever:
* Cyclical fever (every 5-6 days)
* Severe headache, bone pain (especially shins)
* Relapsing, chronic form in some cases

Immunocompromised Patients:
* Bacillary angiomatosis: vascular lesions affecting skin and internal organs
* Culture-negative endocarditis
Chronic Infection:
* Can persist asymptomatically or with mild nonspecific symptoms

Endocarditis
B. quintana is a more frequent cause of endocarditis than B. henselae

Bacillary angiomatosis
Occurs in patients with advanced HIV who have CD4 counts less than 100 cells/microL. It is caused by either B. quintana or B.henselae
¡ Patients with infection due to B. quintana may be more likely to have lytic bone lesions than patients with B. henselae

Bacillary peliosis hepatitis
¡ Also occurs in patients with advanced HIV-related immunosuppression (CD4 <100 cells/microL)
¡ Patients with this syndrome present with abdominal pain and hepatosplenomegaly, with or without systemic symptoms (eg, fever)
¡ Elevated ALP

1399
Q

Who is at risk of Bartonella Quintana infection and how is it transmitted?

A

Body louse (Pediculus humanus corporis)
No known animal reservoir (primarily human-hosted)

WW distribution (US, Europe, Russia, Brazil, East Africa)

Associated with homeless populations and poor sanitation
- HIV, ETOH, IVDU
People in crowded conditions, immunocompromised

1400
Q

How do you diagnose and treat Bartonella Quintana related conditions?

A

DIAGNOSIS
Clinical hx and RFs
Blood culture -> difficult due to fastidious growth
Bartonella serology eg IgM, IgG
PCR
Histopathology for vascular lesions

TREATMENT

Trench Fever:
* Doxycycline (100 mg orally every 12 hours) for 28 days + Rifampicin (300 mg orally every 12 hours) for 14 days
* rule out endocarditis

Bacillary Angiomatosis and Endocarditis:
* Prolonged doxycycline or erythromycin, with gentamicin in severe cases.
* Treatment of BA depends on the presence of bacteremia and severity of illness

1401
Q

Summarise the life cycle of schistosomes

A
  1. Egg Release
    Infected humans release eggs of Schistosoma species through urine (S. haematobium) or feces (S. mansoni, S. japonicum).
    These eggs reach freshwater, where they hatch into free-swimming miracidia.
  2. Miracidia in Snails
    Miracidia penetrate specific species of freshwater snails (intermediate host).
    Inside the snail, the miracidia develop into sporocysts, which produce thousands of free-swimming cercariae over 4-6 weeks.
  3. Cercarial Release
    Cercariae are released from the snail into freshwater. They are free-swimming and infective to humans, their definitive host.
  4. Human Infection
    Cercariae penetrate the human skin during water contact, shedding their tails and transforming into schistosomula.
    Schistosomula enter the bloodstream and travel to the lungs and liver for further maturation.
  5. Adult Worms
    Mature schistosomes migrate to their specific venous locations:
    S. haematobium: Bladder veins
    S. mansoni: Mesenteric veins of the colon
    S. japonicum: Mesenteric veins of the small intestine
    Male and female worms pair and produce eggs.
  6. Pathogenesis
    Eggs released into tissues or excreted cause pathology. Many are excreted into the environment to continue the cycle.
1402
Q

What two key species cause >90% of human schistosomiasis?

What is the general EPI?

A

S.mansoni or S.haematobium

- 78 Endemic countries
- ~800 million at risk
- ~>250 million infected
- 90% of cases are in sub-Saharan Africa
- ~20 million cases of severe morbidity
- ~300,000 deaths per year >90% of cases are S.mansoni or S.haematobium
1403
Q

Describe schistosome development inside snail

A
  • Miracidium infect snail
  • Mother sporocyst in mantle of snail
  • Daughter sporocysts move to snail digestive gland
  • Cercariae develop from daughter sporocyst
1404
Q

Schistosomal egg with lateral spine?

A

S mansoni ~140 microns

1405
Q

Schistosomal egg with terminal spine? Where are the eggs found?

A

S haematobium

Found in urine

~150 microns

1406
Q

What are miricidae in schistosomes aimed for?

A

Penetration of snail host

1407
Q

Correlate of severity in schistosomiasis?

A

Egg production -> eggs and them getting stuck are key causes of pathology

1408
Q

What causes disease in schistosomiasis?

A

Around 50% of eggs are retained in the tissues:-
(A) Intestinal/hepatic species:
(i) In the wall of the intestine
(ii) Carried by the blood and lodged in the liver

(AIM -> to get out of bowel mucosa and be passed out in stool)

(B) Urogenital species: in the walls of the bladder and ureters

(AIM S. haematobium, eggs would pass into the bladder lumen and be released in urine)

Eggs getting stuck triggers immune response -> pathology

1409
Q

A university student goes swimming in the river Cam during a heatwave in the summer and gets a widespread itchy ash. What is this called? What causes it?

A

Cercarial dermatitis (swimmers itch) – commonly caused by animal
or bird schistosome cercariae

Infection with human species generally only give rise to mild dermatitis (if any)

1410
Q

Describe some features of Katayama fever

A
  • 3-8 weeks post infection (onset of egg production). Up to 90% prevalence following first
    exposure, more unusual in endemic settings.
  • Symptoms may include:
    § Malaise, low grade fever
    § Head/neck ache
    § Urticaria
    § Eosinophilia
    § Hepatomegaly, splenomegaly
    § Diarrhoea
    § Cough and wheeze
  • Aetiology uncertain - but probably mediated by immune complexes of soluble worm or egg
    antigens and antibody (hypersensitivity reaction).
1411
Q

What species of Schistosomes cause hepatosplenic schistosomiasis?

A

S.mansoni and S. japonicum

  • Due to eggs getting trapped in pre-sinusoidal vessels of liver
    Granulomatous response, fibrosis and calcification causes periportal fibrosis => portal HTN and hepatosplenomegaly

Also rise in gastric and oesophageal veins -> varices (look for caput medusa)

Risk of acute internal bleeding

1412
Q

Basic pathogenesis of Intestinal/hepatic schisto pathogenesis?

A
  • (Cercarial penetration -> dermatitis)
  • Maturing worms -> Katayama Fever (‘acute schisto’)
  • Escaping eggs -> haemorrhages in bowel mucosa, inflammation, colitis
  • Trapped eggs -> granulomas -> periportal (Symmers’) fibrosis -> Portal hypertension ->hepatosplenomegaly ->oesophageal varices ->haematemesis
1413
Q

How does 90% of schistosomiasis present/manifest?

A

90% chronic anaemia and malnutrition
(leading to fatigue, growth retardation,
loss of cognitive function/development)

1414
Q

Key features and causative species of urogenital schistosomiasis?

A
  • Haematuria
    (Passage of eggs →ulceration of bladder wall → haemorrhage)
  • Fibrosis and calcification of the bladder and ureters
  • Obstructive uropathy
  • Hydroureter, hydronephrosis
  • Bladder cancer
1415
Q

What type of cancer does S.haematobium cause?

A

SQUAMOUS cell carcinoma of bladder

1416
Q

Summarise the pathophysiology of S.haematobium?

A
  • (Cercarial penetration -> dermatitis)
  • Maturing worms -> Katayama Fever (‘acute schisto’)
  • Escaping eggs -> haematuria, proteinuria, bladder polyps and sandy
    patches
  • Trapped eggs in bladder wall -> granulomas -> calcified bladder,
    bladder cancer
  • Trapped eggs in ureters -> calcifications -> hydroureter, hydronephrosis, pyelonephritis
1417
Q

Species that is particularly problematic for neurological schistosomiasis?

A

S.japonicum

~3000 eggs produced per day
Eggs also small

1418
Q

Describe some features of ectopic schistosomiasis

A
  1. Lungs
    • Eggs embolise to pulmonary arterioles -> granulomas ’pulmonary hypertension -> right ventricular hypertrophy and failure (‘cor pulmonale’)
    • Occurs in 15% of S.mansoni Symmers’ cases
    • Also in S.haematobium cases, but rarer in S.japonicum
  2. CNS
    • Eggs ± adults of S.mansoni or S.haematobium in spinal cord -> granulomas -> Paraplegia
    • Eggs ± adults of S.japonicum in brain -> epilepsy etc
  3. Genital tract
    • Up to 75% of women with S. haematobium have eggs in uterine cervix, vagina and vulva and less commonly in uterus, ovaries.
    • GS is associated with “sandy patches”, vascularization and contact bleeding
    • Misdiagnosed as cervical cancer or STIs, GS linked to infertility
    • Women with GS have up to 3-fold increased risk of having HIV
      Men: Eggs common in seminal vesicles, prostate, ~50% of egg +ve men have eggs in semen -> Eggs in scrotal lymphatics →hydrocele, elephantiasis
1419
Q

Clinical features of schistosomiasis?

A

Degree of symptoms will depend on intensity and duration of infection, and
may take years to develop

Acute: fever, diarrhoea, abdominal pain, weight loss

Chronic:
Intestinal/hepatic species: chronic diarrhoea, abdominal pain, anaemia, hepatomegaly, hepatosplenomegaly

Urogenital species: Dysuria, frequency, terminal or total haematuria. Chronic disease – obstructive uropathy, bacteriuria, bladder carcinoma, bladder calcification, genital lesions

Schisto is a protean (changing) multisystem disease with multiple abdominal symptoms (for both types)

1420
Q

Diagnostic techniques for schistosomiasis?

A

PARASITOLOGICAL
identify eggs in:
* sedimented urine, filtrated urine or in semen (Sh)
* Direct smear, Kato Katz or formol-ether faecal concentrates (Intestinal species)

MORBIDITY MARKERS
* Detection of Faecal occult blood (Intestinal species)
* Detection of macro/microhaematuria (Sh) (hemastix)

IMMUNOASSAYS
* Eosinophilia (~45% of patients)
* Detection of specific antibody (e.g. to soluble egg antigens by ELISA) (but time to seroconversion
often several months)
* Detection of antigen in urine (e.g. ‘circulating cathodic antigen’(CCA) by RDT) -> only for S.mansoni

1421
Q

What schisto species can be diagnosed with a RDT?

A

S.mansoni

Urine CCA antigen (RDT) -> “circulating cathodic Ag” -> semi-quantitative assessment of adult worm burden

1422
Q

Treatment and issues with treatment of schistosomiasis?
New development of treatment?

A

40-60 mg/kg p.o, single dose

Issues with PZQ
* Side effects: abdominal cramps, nausea, diarrhoea, dizziness, vomiting – worse with higher intensity
infections

  • it cannot be used for chemoprophylaxis due to its short half-life, and it is ineffective against
    migrating schistosomula and juvenile worms
  • Longstanding availability problems: in 2019 only 44.5% of people requiring treatment globally
    were reached (WHO) (in 2021 only 29.9% were reached due to the pandemic)

NEW
- Paediatric formulation coming out which tastes less bitter, water soluble and is smaller

1423
Q

Describe the epi, intermediate hosts transmission of Schistosomiasis and location within human host of worms (according to key species)

A

Transmission sites: Lake, streams (slow flowing), irrigation systems, dams, rice fields

S.haematobium
- Africa, Middle East
- Snail: Bulinus spp
- In human: Small venules of the vesical plexus and pelvic plexus and sometimes also the rectal venules

S.mansoni
- Africa, South America, Caribbean
- Snail: Biomphalaria spp.
- In human: Mesenteric veins of small and large intestine

S.japonicum
- Southeast Asia (China, Philippines, etc.)
- Snail: Oncomelania hupenis (water and mud)
- In human: Mesenteric veins of the small and large intestine (but can be anywhere in mesenterics)

1424
Q

Describe 5 prevention techniques for schistosomiasis

A
  • Health Education
  • Safe water supplies
  • Sanitation
  • Snail control (by habitat modification
    and/or molluscicides)
  • Chemotherapy campaigns
1425
Q

What are schisto MDA programmes designed for and what are some issues with them?

A

MORBIDITY CONTROL
using praziquantel to prevent build-up of severe fibrosis in
individuals living in endemic areas (primarily school age children)

Issues
- Risk of reinfection high due to
- Lack of effect of PZQ on juvenile worms -> individuals still infected and will continuing shedding eggs -> transmission ongoing
- High levels of infection prevalence and intensity
- Poor or non-compliance of PZQ treatment
- Low coverage
- Recontact with contaminated water as result of daily activities and seasonal factors

1426
Q

WHO 2022 guidelines on control and elimination on human schistosomiasis

A
  1. Endemic communities with prevlance of S.spp. >/=10% ANNUAL preventive chemotherapy with single dose of praziquantel at >75% treatment coverage in all age groups 2+ years old

(includes adults, pregnant women after the first trimester and lactating women)

  1. WHO recommends WASH interventions, environmental interventions (water
    engineering and focal snail control with molluscicides) and behavioural change as essential methods to reduce transmission
1427
Q

Describe the egg morphology of the 3 disease causing schistosomes

A

S. haematobium
- Seen in URINE
- Terminal spine
- 150 microns

S.mansoni
- See in stool sample
- Lateral spine
- 140 microns
- Production: ~300 eggs per day per female

S.japonicum
- Small lateral spine, often hard to see
- 85 microns
- Production: ~3000 eggs per day per female

1428
Q

What is Clostridium perfringens associated with?

A

Gas gangrene

Severe haemolytic anaemia

1429
Q

What is chorioretinitis?

A

Inflammation of choroid (vascular layer) and retina

Clinical Features:
Visual Symptoms: Progressive reduction in visual acuity or visual field loss, often more severe than anterior uveitis.

Painless: Usually no pain unless there is secondary involvement of other eye structures.

Floaters: Patients may report seeing spots or shadows due to inflammatory cells in the vitreous.

Fundoscopic Findings:
Yellowish-white lesions in the retina and choroid, with potential scarring in chronic cases.
Retinal hemorrhages or vascular sheathing in severe inflammation.

1430
Q

What is sclerosing keratitis?

A

Chronic inflammation of cornea -> characterised by scarring and fibrosis

Progressive Vision Loss: Typically from peripheral corneal opacification extending toward the central cornea.

Photophobia: Sensitivity to light due to corneal involvement.

Pain: Mild to moderate eye discomfort due to corneal inflammation.

Physical Exam:
Opacification or whitening of the cornea, typically starting in the peripheral cornea.

In severe cases, vascularization of the cornea (blood vessels invading corneal tissue).

1431
Q

How can you help prevent neonatal tetanus via vaccination?

A

Tdap-IPV vaccine is a combined inactivated vaccine that protects against tetanus, diphtheria, acellular pertussis, and polio

Vaccinate pregnant women between 16 and 32 weeks gestation (ideally fetal anomaly scan at 20 weeks)

1432
Q

What is the positive predictive value?

A

Proportion of those that test positive that genuinely have the disease

  • increases with higher prevalence
  • increases the more specific the test
1433
Q

What is sensitivity?

A

Proportion of people who have the disease that are correctly identified by the test

1434
Q

A 70-year-old man had blood test from his family doctor as part of investigations for possible diabetes. He was well. He had spent much of his adult life until the age of 50 working in sub Saharan Africa and Southeast Asia. He had not left the UK in the last 20 years. His eosinophil count is 0.94. What is the most likely parasite?

Ascaris lumbricoides
Giardia lamblia
Trichuris trichuria
Schistosoma mansoni
Strongyloides stercoralis

A

Strongyloides stercoralis

1435
Q

EPI of TB

A

70% of global TB burden in 8 countries
1. India (27%)
2. Indonesia (10%)
3. China (7%)
4. Philippines (7%)
5. Pakistan (6%)
6. Nigeria (5%)
7. Bangladesh (4%)
8. DRC (3%)

1436
Q

What NCD should people with TB be screened for?

A

Diabetes

1437
Q

Rash with Lyme

A

erythema chronicum migrans

1438
Q

Most common cause of meningitis in UK?

A

Neisseria meningitidis group B

1438
Q

Causes of meningitis in meningitis belt?

A

Neisseria meningitidis group A

1439
Q

Staph. epidermidis micro features

A

gram positive cocci in bunches

associated with line infections but can also be floral skin contaminant

1440
Q

In what cases may TST be negative?

A

Young children, the elderly and the immunosuppressed may fail to mount an adequate immune response. They are often tuberculin negative in the presence of active disease. In such cases the patients often become tuberculin positive during treatment.

can be positive in
- tuberculosis pericarditis
- in 30% of sarcoid cases

1441
Q

usually diagnosed by microscopy of adhesive tape prints taken from the perianal area?

A

threadworm = pinworm
enterobius vermicularis

1442
Q

What nematode infection may block the pancreatic duct causing pancreatitis?

A

Ascaris lumbricoides

1443
Q

What are factors associated with an increased risk of vertical transmission of HIV?

A

high p24 Ag
low CD4
breast feeding
HIV-1 (compared to 2)

1444
Q

Incubation period of African trypanosomiasis?

A

10-21 days
from time of bite to chancre

1445
Q

Important causes of zoonotic infections?

A

Viral
orf
cowpox
rabies
arboviral infections

Bacterial
salmonella
campylobacter
brucella
tuleraemia
Weil’s disease
anthrax

Protozoal
African trypanosomiasis
Leishmaniasis
giardiasis
toxocariasis
cryptosporidiosis

1446
Q

A fever of two weeks’ duration associated with neutropaenia is characteristically due to

A

malaria
brucellosis
typhoid
disseminated tuberculosis
visceral leishmaniasis

1447
Q

Causes of Erythema nodosum?

A

Streptococcal infection
Tuberculosis
Sarcoidosis
Leprosy
Sulphonamides
Inflammatory bowel disease

1448
Q

Coagulopathy may follow envenomation by which snake?

A

African pit vipers

1449
Q

What happens to your microvasculature in dengue? How does this manifest?

A

The micro-vasculature is affected by a number of things:
1. Plasma leak
2. Endothelial permeability
3. Thrombocytopenia and bleeding

Narrow pulse pressure

1450
Q

Vector control methods for dengue?

A
  • Environmental management
    – Elimination of container habitats:
  • Piped water systems
  • Mosquito-proofing water-storage containers
  • Disposal of waste (water-bearing containers)
  • Chemical control
    – Larvicides & insecticides
    – Indoor Residual spraying
    – Space spraying/fogging (emergency measure)
  • Biological control: Wolbachia
1451
Q

Grading and relevant features of neonatal encephalopathy?

A

Mild
- Hypertonic
- Hyperalert, starring
- Partial primitive reflexes
- Irritable, hand clenching
- Voracious/poor feeders
Moderate
- Hypotonic
- Lethargic
- Don’t feed
- Absent primitive reflexes
- Strong distal flexion
- Apnoeas
- Seizures
Severe
- Completely floppy
- Comatose
- Apnoeic
- Decerebrate posturing
- Absent reflexes
- Full fontanelle

1452
Q

Manifestations of seizures in babies and management

A

In babies: eye deviation, flickering of eyes, stereotypical mouth movement, cycling/regular twitching

Management:
1) phenobarbitone: loading, starting with 20mg/kg to total 40mg/kg
2) phenytoin 18mg/kg

1453
Q

What is cerebral palsy and what are the different types?

A

Types
Quadriplegia: 4 limbs affected
Diplegia: lower limbs affected
Hemiplegia: arm and leg on one side affected

- Spastic: high tone, tight rigid muscles
- Hypotonic: low tone (loose muscles and joints)
- Athetoid: fluctuating muscle tone and uncontrolled movements
- Ataxic: lack of coordination Mixed: combo of 2 or more
1454
Q

Tools for screening/assessment of children with developmental disorders including CP

A
  1. Global scales for early development: open access package -> standardized method for measuring development of children up to 36months, creates developmental score (D-score) using short form and long form
  2. WHO caregiver skills training: focused largely on autism and behavioural disorders for 2+ year olds, no content on CP

Issues:
- Stigma, exclusion & discrimination
- Gender, equality and empowerment
- Poverty and livelihoods
- Literacy, language, fidelity

1455
Q

What is Keratitis?

A

Inflammation of the cornea

1456
Q
A
1457
Q

Management of measles?

A
  • Systemic management (antibiotics; rehydration etc)
  • High dose vitamin A x 3 doses [200,000 IU orally, or IM]*
  • Examine the eyes, even if child very sick with eyes closed (VAD is painless)
  • Use fluorescein stain to detect ulceration
  • Topical antibiotic e.g. tetracycline ointment
  • Urgent referral to eye unit if concerned about the eyes
    *Saves sight and lives
1458
Q

What are the stages of retinopathy of prematurity? (in general, no details)

A

6 stages: 1/2 usually improve and watch + wait, stage 3+ treat, stage 4: retinal detachment, stage 5: total detachment, stage 6: blind

1459
Q

What are some important factors for disease manifestation of helminth infections?

A
  • Number of worms infecting the host (related to level of exposure, cumulative after repeated exposures)
  • Duration of infection (long life span of worms)
  • Immune response to infection (may cause immune-mediated pathology)
  • Location of worms (migration?)
  • Age of host (childrens habits make them more likely to be exposed but they may also suffer more from
    disease, anemia etc)
  • Sex of host (women may suffer more from helminth-induced anemia)
1460
Q

What are the 3 key ‘types’ of nematode?

A

Intestinal: direct life cycle, soil-transmitted *eggs need contact with soil to become infective, ?for embroynation but not well understood

Filarial: indirect life cycle involving insect or copepod

Zoonotic:
nematodes of animals that occasionally are transmitted to humans -> humans often dead-end host

1461
Q

What intestinal nematode is NOT soil transmitted?

A

Enterobius vermicularis

1462
Q

What are the key intestinal nematodes and how are they transmitted?

A

(i) in/on the soil (geohelminths, STH)
- Trichuris trichiura (oral)
- Ascaris lumbricoides (oral)
- Hookworms: Necator americanus, Ancylostoma duodenale (percutaneous)
- Strongyloides stercoralis (percutaneous)

(ii) without needing contact with soil
- Enterobius vermicularis (oral)

1463
Q

What are the WHO recommended drugs for MDA for soil transmitted helminths? What’s the normal prevalence cut off for this?

A

Albendazole
Mebendazole

Levamisole
Pyrantel

Prevalence >20%

1464
Q

What is the most common
helminth infection in the USA and Western Europe?

A

Enterobius vermicularis = pinworm/threadworm

1465
Q

Diagnosis and treatment of enterobius vermicularis?

A

DIAGNOSIS
Cellotape test (before showering in mornings, ideally x3)

TREATMENT
Single dose mebendazole or albendazole (kills adults)

Repeat after 2-4 weeks (due to resistant eggs and risk of re-infection), treat whole family

1466
Q

Epi and control/prevention of Enterobius?

Life cycle?

A

Cosmopolitan, high and low income countries
Playgroups, nurseries, crowded conditions etc
Most common
helminth infection in the USA and Western Europe
~300 million cases worldwide
Prevalence up to 40% in US children

Oral (fecal-oral, autoinfection)
NOT soil transmitted
Found small -> large intestine (cecum, appendix)

LIFE CYCLE
Ingestion -> infective within hours
Eggs hatch in small intestine + larva move to large intestine
Penetration and development in mucosa
Adult maturation (2-6 weeks)
Males die
Gravid females migrate to perianal region to lay eggs at night

CONTROL
Anti-helminths
Hygiene, cleaning, washing, clothes, bed linen

1467
Q

Describe the site of infection and life cycle of trichuris?

Control measures?

A

SITEs
Epithelium of large intestine (cecum, colon) -> no migration anywhere else
Can have adults in rectum and terminal ileum in heavy infection

LIFE CYCLE
Eggs hatch and produce larvae in small intestine
Larvae migrate to colon to mature and embed in mucosa
Release unembryonated eggs in stool ~8 weeks after infection
Adults live >2 years

CONTROL
Sanitation
Ingestion can occur due to night soil ie use of human faeces as fertiliser then consuming unwashed vegetables
WHO recommend mass school-based chemotherapy campaigns if prevalence in the area >20%

1468
Q

Kato Katz thick smear is recommended for diagnosing what infection?

A

Trichuris trichuria

1469
Q

Clinical features, diagnosis and treatment of trichuris infection?

A

CLINICALLY
Light -> asymptomatic
Mod/heavy -> oedematous, haemorrhagic mucosa “chronic Trichuris colitis” = Trichuris dysentery syndrome
- Risk of bacterial infection with Shigella and Salmonella
- Risk of invasion by E.histolytica

Chronic: sub-clinical effects on physical and cognitive development in children

Worms burrow into intestinal epithelium -> feed on cells -> small haemorrhages + inflammation

DIAGNOSIS
Eggs in stool
- Direct smear
- Kato Katz thick smear (WHO recommended)
- Formol-ether concentration
- FLOTAC

TREATMENT
Albendazole or Mebendazole 100mg BD 3 days
Or
Oxantel + pyrantel

1470
Q

ASCARIS LUMBRICOIDES
- epi
- transmission
- site
- life cycle

A

EPI
- Columbia/Venezuela
- West/Central Africa eg Cameroon, Congo, Nigeria
- Kazakhstan
- South East Asia
- Papua

TRANSMISSION
Oral (fecal-oral)

SITE
Larval lung migration before adults settle in small intestine

LIFE CYCLE
Ingest eggs
Eggs hatch in duodenum but do not directly develop -> L3 larvae penetrate gut mucosa -> lymphatics or veins -> liver -> heart -> lungs ->
penetrate alveoli
ascend trachea
Swallowed
Develop into adults in gut

Eggs (females produce ~200,000 a day) released into faeces -> very resistant to desiccation so can survive in environment for 8 years

Eggs in stool 9-10 weeks after ingestion
Adult worms live 1 year on average

1471
Q

Ascaris lumbricoides
- 3 phases?
- diagnosis?
- treatment?

A

3 phases
1. Lungs
○ Loeffler’s: dry cough at 1-3 weeks, dyspnoea, asthma, substernal pain, haemoptysis, eosinophilia
2. Intestinal
○ Abdo discomfort, acute colicky pain, poor sleep, diarrhoea, malabsorption -> malnutrition
○ Intestinal obstruction -> bolus of worms stuck at ileo-caecal valve
○ Risk of necrosis, gangrene, death
3. Ectopic
○ Migration of adults
○ Promoted by fever, anaesthetics, antihelminth tx
○ Appendix
○ Pancreas -> haemorrhagic pancreatitis
○ Bile ducts
○ Peritoneal cavity
○ Larynx, lungs, urethra, vagina, heart, brain etc

DIAGNOSIS
Eosinophilia in ~10%
Stool microscopy: knobby-surfaced eggs
Can also use Kato-Katz preparation

TREATMENT
Albendazole, Mebendazole, or pyrantel

Benzimidazoles cause flaccid paralysis of adults reducing risk of obstruction

1472
Q

How do you distinguish between L1 rhabditiform larvae of Hookworm vs Strongyloides?

A

Hookworm = long buccal invagination

Strongy = short buccal invagination

1473
Q

Hookworm
-EPI
- Transmission
- Site of transmission

A

Warm moist climates -> limited by humidity and temp requirements (~23-28 degrees) of infective stages

Necator more in Latin America, central/SS Africa and SE Asia

Ancyclostoma in SE Asia and North Africa

Larvae in environment need moisture, warmth, shade, decaying vegetation

Percutaneous (skin penetration by larvae) - often feet
Or
Oral ingestion of unwashed vegetation (Ancyclostoma>necator)

Small intestine
Exhibit blood feeding

1474
Q

Features of L3 larvae of hookworm

A

retains sheath of L2
sharply pointed tail
won’t see full larva in 40x magnification

1475
Q

Describe the life cycle of hookworm

A

L3 filariform larva penetrates skin
L3 migrate through alveoli-> trachea -> swallowed to intestine
L3 -> L4 -> adults which bite onto mucosa of small intestine
Eggs in faeces
L1 Rhabditiform larva hatches + feeds on bacteria in stool
L2 also feeds
L3 filariform larva = infective (after 8-10 days), does not feed, lives a few weeks

Egg laying around 5-8 weeks post-infection

If infection with L3 is oral -> heart-lung migration

1476
Q

What are the clinical manifestations and pathophysiology of hookworm infection?

A

In general: weakness, lethargy, abdominal discomfort, pallor, anaemia, eosinophilia in acute phase

3 organs

  1. Skin
    ○ 1-2 weeks post-exposure
    ○ L3 larval penetration
    ○ CML “cutaneous larva migrans” = Serpinginous, erythematous rash, may move 2-5cm/day, can last a few weeks
    ○ Severity of itch depending on no. of penetrating larvae
  2. Lungs
    ○ 1-2 weeks post-exposure due to L3 migration into alveoli
    ○ Pneumonitis, bronchitis, pharyngitis
    ○ Dry cough, wheezing
    ○ Related to severity of infection
  3. Intestine
    ○ Blood loss due to bite
    ○ Hookworm pump blood through bodies to feed on erythrocytes
    ○ Worms move frequently -> persisting haemorrhages due to release of anticoag of worms
    ○ Heavy infections -> >100ml blood loss/day

Long term: cardiac insufficiency, physical and mental stunting due to Fe-deficiency anaemia
- More likely chronic than acute, ie due to gradual accumulation
- Dependent on inadequate Fe intake (common in most endemic areas)
- Anaemia often compensated but upset by slight changes in Fe reserves/intake eg malaria, schiso, menstruation, pregnancy

Also get hypoalbuminemia + resulting oedema

Can get weight loss due to malabsorption

May be fatal at high intensity, esp in children

Blood loss and anaemia directly proportional to worm burden

1477
Q

Diagnosis and treatment of hookworm?

A

DIAGNOSE
Stool microscopy
- Fresh: eggs
- Old eg >24hrs: hatched L1 larvae
Anaemia on FBC

TREAT
Albendazole, Mebendazole; iron supplementation

CONTROL
- avoid walking barefoot in contaminated soil etc
- WHO recommends MDA if prevalence >20%

1478
Q

Clinical manifestations of
- strongyloidiasis in immunocompetent host
- strongy in immunosuppressed host (+ relevant organs affected)

A

Asymptomatic

Skin: itchy dermatitis, i) non-specific urticarial rash of crops of stational wheals lasting 1-2 days OR ii) larva currens (10cm/hr) ++ itchy

Lung: pneumonitis, asthma/allergy s/s, cough, CP (sometimes mistaken for asthma, patients given steroids and then get disseminated disease)

Intestine: mucosal damage leading acutely to upper abdo pain mimicking peptic ulcer, diarrhoea, blood/mucus in stool with ++ eosinophilia, chronic: asymptomatic, sometimes chronic colitis, haemorrhage, malabsorption (IgE raised, eosinophilia lower)

Eosinophilia, abdominal pain, diarrhea, hyperinfection syndrome in immunosuppressed (can be fatal)

Disseminated strongyloidiasis
- Not just chemo/severe malnutrition/lymphoma etc
- Now due to widespread use of corticosteroids + HTLV-1
- NB: HIV NOT associated with DS
- Sometimes treat in absence of positive test based on travel hx + eosinophilia

Lung: cough, dyspnoea, wheezing, resp distress + death from resp failure +/- pul haemorrhage

Intestine: parasites damage submucosa -> oedematous mucosa/submucosa, mucosal/villous atrophy, sprue like syndrome: steatorrhea, protein losing enteropathy, weight loss, paralytic ileus
Ulceration + bacteria carried by larvae -> gram neg bacterial infection -> peritonitis due to bowel perforation, septicaemia, secondary bacterial infections of CNS (brain abscess + meningitis)

1479
Q

Strongyloides stercoralis
- Epi
- Transmission
- Site

A

EPI
Worldwide but most common in warm moist climates
overlapping with hookworm distribution; sporadically in Europe, N. America and Japan

TRANSMISSION
Percutaneous (skin penetration)

SITE
Small intestine

1480
Q

Life cycle strongyloides stercoralis

A

1-4 days in soil

Direct life cycle:
L3 rhabditiform larvae penetrates skin, undergoes heart-lung migration
Swallowed -> small intestine
Females reproduce parthenogenetically (without males)
L1 rhabditiform larvae release

Indirect:
L1 rhabditiform larvae released in stool from human host
Develop into free living adult males and females
Adults mate in stool -> eggs -> hatch into new L1 larvae
L1 larvae can either
- Independently develop into rhabditiform larvae to continue free-living cycle
- OR develop into L3 filariform larvae

Self infection:
Precocious development of L1->L3 within gut
L3 invasion of gut mucosa
Internal autoinfection
L3 in faeces contaminating perianal regions or following development of L1->L3 on perianal skin can penetrate same host
= external autoinfection
L3 undergo heart-lung migration -> replicating infection
Occurs at low level in normal host -> persisting infections last >50 years

In case of severe cellular immunodeficiency -> loads of larvae autoinfect -> adults in gut “hyperinfection syndrome” + disseminated strongyloidiasis (lots of l3 larvae cross capillary bed of lungs to infect all organs)

1481
Q

Diagnosis and management of strongyloides?

A

DIAGNOSITICS
Stool microscopy with serial examinations (due to intermittent larvae excretion)
Direct faecal smear* or concentration methods: formol-ether, baermann
- Fresh: Observe L1 larvae

Culture: charcoal*, nutrient agar plate

Baele’s string test

Duodenal aspirate

Disseminated disease: larvae in sputum and sometimes urine

Immunodiagnostics:
ELISA* with L3 Ag

TREATMENT
Ivermectin (first line)
Albendazole (alternative)

Repeat treatment as migrating larvae insusceptible to drugs (+ re-treat immunosuppressed patients)

1482
Q

Strongy
- complications of hyper infection
- triggers for hyper infection

A

Complications
- Larva currens (relatively straight)
- Nephrotic syndrome
- ARDS
- Recurrent gram negative bacteraemias (and can’t establish source)
- Chronic diarrhoea
- Bowel obstruction

Triggers:
- High dose steroids
- HTLV-1
- Transplant

1483
Q

Cavitating lesion on CXR - what is acronym for causes?

A

TANKS CRAP MD

TB
Aspergillus
Nocardia
Klebsiella
Staph/strep

Cryptococcus
Rodococcus (associated with soil around horse stables)
Actinomycosis (can cause mycetoma)
Paragonimus

Mucor, melioidosis
Dimorphic fungi

1484
Q

Differentials for bipolar staining - safety pin? (remember acronym)

A

BuY PaT
- Burkholderia
- Yersinia
- Pasturella
- Tuleraemia

1485
Q

3 key causes of eosinophilic meningitis?

A

BAG
Bayliscaris procynosis (old racoon poo)

Angiostrongylus cantonensis (eat slugs or snails or vegetables they have been moving across)

Gnathostoma spinigerum (raw fish)

1486
Q

WHO management of acute severe malnutrition?

A

Treat or prevent
1. Hypoglycaemia
2. Hypothermia
3. Dehydration
4. Correct electrolyte imbalance
5. Treat infection
6. Correct micronutrient imbalance
7. Begin feeding
8. Increase feeding to recover lost weight (catch up growth)
9. Stimulate emotional and sensorial development
10. Prepare for discharge

At a later date then treat helminth infections

1487
Q

Time of day for Loa Loa blood film?

A

Daytime
- Loa loa comes out at lunch

1488
Q

When do you do a blood film for LF?

A

At night

1489
Q

Intermediate host for S.haematobium?

A

Bulinus snail

1490
Q

What are the two drugs for Loa Loa and how do they work?

A

DEC: diethyl carbamazine targets macrofilaria

Doxy: targets microfilaria

1491
Q

Facts about Ebola transmission/epidemic

A

Many come from single source eg patient

Ring vaccination is effective: contacts and contacts of contacts

Babies: test before breast feeding and wear full PPE etc when delivering baby to Ebola mother

Infective Ebola virus can be found in various fluids for weeks to months after
recovery

The high fatality rate was from multi-organ failure and shock rather than from haemorrhage

1492
Q

Lymphatic filariasis
- Epi
- Causes
- Transmission

A

Epi

India, SE Asia, Pacific Islands, West Africa and Brazil
Brugia malayi: India and SE Asia

Causative Agents

Wuchereria bancrofti (90% of cases)
- Humans definitive host
- Cosmopolitan
Brugia malayi
- Periodic and sub-periodic strain
- Periodic: mansonia (vector), humans definitive host
- Sub-periodic: range of mosquito vectors, range of hosts eg humans, monkeys, domestic casts, forest carnivores
Brugia timori

Vector transmission

Transmitted by mosquitoes: Culex, Anopheles, Aedes, Mansonia
Periodicity of larvae -> nocturnal (come out at night due to vector biting habits)
- Nocturnal: Wuchereria bancrofti, Brugia malayi
- Nocturnal sub-periodic: Brugia malayi
- Diurnal periodic: Wuchereria bancrofi, pacific islands

Reverse in pattern of periodicity in places where day biting of mosquitoes eg Aedes polynesiensis (in Pacific Islands), bites during day so filariae come out during day

1493
Q

Life cycle lymphatic filariasis

A

Infected mosquitoes deposit L3 larvae (which have developed over 2 weeks)
Mature into L4 in lymphatics in 2 weeks
Mature into adult worms (male and females) in 6-12 months and live in lymphatics (live for ~10 years)
Adult worms produce microfilariae that circulate in blood
Mosquitoes ingest microfilariae, move to midgut
Shed sheath
Get into flight muscle cells and moult into L2
Grow then develop into L3 infective larvae in these cells after 10 days

1494
Q

Clinical presentation and grading of LF?

A

Acute phase: Fever, skin rashes, lymphangitis, lymphadenitis, epididymoorchitis
- Due to death of parasite in lymphatics -> inflammatory reactions
Dematolymphangioadenitis stage: constant episodic inflammation +/- secondary bacterial infection, pain, disability
Chronic phase: Elephantiasis (limb and genital swelling), hydrocele, chyluria
Tropical Pulmonary Eosinophilia (TPE): Cough, wheezing, eosinophilia

Affects children + damages their lymphatics but only see clinical manifestations as adults

Grade:
- Stage 2: unreversible swelling
- Stage 3: shallow skin fold - Stage 4: alternation of skin texture, formation of knobs
- Stage 5: deep skin folds
- Stage 6: mossy lesions
- Stage 7: inability to perform work

1495
Q

Role of Wolbachia in LF?

A

These mutualistic endosymbiotic bacteria are essential for:
Development
* Larval development
* Embryogenesis
PROPHYLAXIS
TRANSMISSION BLOCKING

Disease pathogenesis
* Inflammatory reactions
* Clinical disease
CLINICAL CASE MANAGEMENT

Adult worm survival (10-14 years)
* Adults depleted of Wolbachia
die 1-2 years later
MACROFILARICIDAL (kills adult worms)

1496
Q

Diagnostic techniques for LF

A

Diagnosis

Thick blood smear for microfilariae (best collected at night due to periodicity)

  • Wuchereria: nuclei do NOT extend to tip of tail
  • Brugia: two small terminal posterior nuclei

Antigen detection (Immunochromatographic Test for W. bancrofti): IgG4 Ab to BmR1

  • But cross reaction with Loa Loa
    Ultrasound (filarial dance sign)
  • Helpful to do prior to treatment to “map” where worm nests are then repeat after treatment
  • Eosinophilia in TPE
1497
Q

Common bacteria causing dental/oral infections

A
  • “Viridans”
    Streptococci–S.mitis,
    S.salivarius,
    S.mutans
  • Oral anaerobes including Actinomyces sp.
  • Fusobacterium
  • S.aureus
1498
Q

Common bacteria causing URT/ENT infections

A
  • Streptococcus pneumoniae
  • Streptococcus pyogenes
  • Haemophilus influenzae
  • Mycoplasma pneumoniae
  • Moraxella sp
  • Chlamydia pneumophilia
  • Fusobacterium necrophorum
  • Neisseria meningitidis
  • Pseudomonas aeruginosa
1499
Q

Common bacteria causing URTIs?
Any others for COPD patients?

A

Common
* Streptococcus pneumoniae
* Haemophilus influenzae
* Mycoplasma pneumoniae
* Legionella pneumophila
* Chlamydia pneumoniae

If hx COPD
* as above plus:
Pseudomonas aeruginosa

If aspiration, as above
plus:
* Anaerobes
e.g. Bacteroides sp., Fusobacterium sp

Zoonotic:
* Chlamydia psittaci

If empyema:
* Staphylococcus aureus
Streptococcus pneumoniae
Streptococcus anginosus group

Others
* Mycobacteria

1500
Q

Common bacterial causes of skin and soft tissue infections?

Some post-op causes?

A

Staphylococci sp.
–esp S.aureus
* Strep.pyogenes
* Group C/G streptococcus
* Cutibacterium sp.
* Corynebacterium sp.

Post-op
* Pseudomonas
* Enterobacterales

1501
Q

Common bacterial causes of skin and soft tissue infections from bites

A
  • Oral anaerobes
  • Staph aureus
  • Streps
  • Neisseria sp
1502
Q

Goals for NTDs 2021-2030 to achieve SDGs? (4)

A
  • To reduce by 90% the number of people requiring treatment for NTDs
  • To eliminate at least one NTD in 100 countries
  • To eradicate two diseases (dracunculiasis and yaws)
  • To reduce by 75% the disability-adjusted life years (DALYs) related to NTDs

The new road map seeks to promote a paradigm shift in the way NTDs are
addressed by;
health.
1) Moving away from process driven targets to measures of the impact on public
2) Replacing vertical and siloed disease specific programmes with holistic cross-
cutting approaches integrated into national health systems with a people and
community centred focus.
3) To transfer programme ownership away from partner support and reliance on
donor funding to country ownership with NTDs integrated into national health
plans and budgets.

1503
Q

Common bacterial infections of adults vs kids for bones and joints?

Others for prosthetic joints?

A

Adults:
* Staph aureus
* Strep.pyogenes
* Group C/G streptococcus
* Strep pneumoniae
* Neisseria gonorrhoea

Kids:
* Staph aureus
* Haemophilus influenzae
* Strep pyogenes
* Strep pneumoniae
* Kingella kingae

Prosthetic joints:
* Staph aureus
* Coag neg staph
* Corynebacteium
* Pseudomonas
* Enterobacterales
* Cutibacterium

1504
Q

Common bacterial causes of GI tract infection?

A

Common
* E. Coli
* Shigella
* Campylobacter sp
* Klebsiella sp.
* Pseudomonas aeruginosa
* Bacteroides fragilis
* Strep. anginosus/constellatus/intermedius
* Clostridium difficile
* Helicobacter pylori
* Salmonella

Less common
* Vibrio cholerae
* Listeria monocytogenes

1505
Q

Causes of bacterial endocarditis?

A

Common
* Staphylococci
–S.aureus
–Coagulase neg. Staph
* Oral streptococci
* Streptococcus gallolyticus
* “HACEK”
* Enterococcus sp.

Less common
* Coxiella
* Bartonella
* Brucella

1506
Q

Common bacterial causes of genitourinary tract?

A

Common:
* E.coli
* Klebsiella sp.
* Staphylococcus
saprophyticus
* Enterococcus sp.
* Proteus sp.
* Strep.agalactiae (GBS)

Other:
* N.gonorrhoea
* Chlamydia trachomatis
* Treponema pallidum

1507
Q

Common sources of brain abscesses?

A
  • Skin
  • Dental
  • Ear / mastoid / sinuses
  • Lung
  • Gut
  • Heart valve

Other:
- TB
- Nocardia

1508
Q

Common bacterial causes of meningitis?

A

Common
* Neisseria meningitidis
* Streptococcus pneumoniae

Elderly, Pregnant
* As above plus:
Listeria monocytogenes
Unvaccinated
* Haemophilus influenzae B

Neonates
* Streptococcus agalactiae
* Listeria monocytogenes
* Escherichia coli
* Klebsiella sp.
* Serratia marcesens

CNS instrumentation
* Skin flora
* Pseudomonas
* Gram negatives

Other
* M.tuberculosis

1509
Q

Potential anti-Wolbachia drugs?

A

Current licensed: Doxy

Other candidates:
- High dose Rif
- Rif + Albendazole

Other proof of concept drugs:
- Rif
- Rifapentine
- Moxifloxacine
- Minocycline

1510
Q

Lymphatic filariasis
- Treatment
- Prevention
- Public health programmes

A

TREATMENT

Africa: Albendazole + Ivermectin
Asia/Americas: Albendazole + DEC
- Single dose, annual 2 drug regimen, mass treatment of at-risk populations + interrupts transmission

Alternative:
- DEC or DEC + Ivermectin for 4-6 years
- DEC fortified salt

Diethylcarbamazine (DEC): kills both adult worms and microfilariae.
Ivermectin: Microfilaricidal.
Doxycycline: used to deplete Wolbachia but need to kill >90% of Wolbachia otherwise doesn’t work

Enhanced self care includes deep breathing exercises, elevation of leg, hygiene for lymphoedema and Abx/anti-inflammatory in acute attacks

PREVENTION

Vector control (insecticide-treated nets, removing breeding sites)
Mass Drug Administration (MDA)
Africa: Albendazole + Ivermectin
Asia/Americas: Albendazole + DEC
- Single dose, annual 2 drug regimen
MMDP = morbidity management and disability prevention runs alongside MDA +/- hydrocoelectomy (surgical inversion of tunica vaginalis)
Personal protection measures like repellents

PUBLIC HEALTH PROGRAMMES

GPELF - Global Programme to Elimination of LF launched in 2000, changed to GAELF (Global alliance to eliminate LF)
Two key strategies:
1. To stop the spread of infection (MDA)
- New WHO strategy with triple therapy IDA = Ivermectin, DEC, Albendazole (as standard tx outside of Africa)
- Get sustained suppression of microfilariae for 3 years, so should only need 2x MDA every 3 years
2. To alleviate the suffering (MMDP = morbidity management and disability prevention)
- Need six annual treatments with Ivermectin, Albendazole, DEC (these suppress microfilariae for duration of life of adult females, until infection dies out)

A.WOL: Aim: to deliver an oral anti-Wolbachia drug effective in ≤7 days
Current licensed: Doxy

Other candidates:
- High dose Rif
- Rif + Albendazole

Other proof of concept drugs:
- Rif
- Rifapentine
- Moxifloxacine
- Minocycline

1511
Q

Differentials for (infectious) retinitis?

A

BACTERIA
Syphilis
Bartonella
Brucella
Coxiella
Endocarditis
whipples

VIRAL
CMV
HSV
VZV
HIV
SSPE

PARASITIC
Toxocara sp
Toxoplasmosis gondii
Baylisascaris
procyonis
Gnathostomiasis
Cystercisosis
Onchocerciasis
Loiasis etc.

MYCOBACTERIAL
Tuberculosis

FUNGAL
Histoplasmosis sp
blastomycosis sp
coccidiomycosis sp
Candida
cryptococcosis

NON-INFECTIVE
Sarcoid
connective tissue
disorders, vasculitis,
lymphoma/leukemia
drug induced
pregnancy related etc.

1512
Q

Where is Baylisascaris found?

A

Japan, Canada, Parts of Europe

1513
Q

Where is gnathostoma found?

A

High cases in Thailand

1514
Q

Where is angiostrongylus?

A

Rat lung worm
Eyes and brains -> neurotropic

America
Vietnam
Thailand

1515
Q

Pathophysiology of rickettsial diseases?

A

Eschar - preceding symptoms

Fever - at day 0/1

Rash - day 4

NB: no Abs in first week, can only get seroconversation after 14 days = GOLD STANDARD Immunofluorescence assay for these infections (IgM and IgG)
To diagnose acutely: need PCR

1516
Q

Gold standard test for rickettsial diseases?

A

Indirect immunofluorescence IgG
antibody assay of paired serum samples.

1517
Q

Common features of all rickettsial diseases?

A
  • All spread by arthropods
    Vectors: fleas, mites, lice, ticks
  • Often also act as reservoir
    Reservoir: small mammals / birds
  • Present all around the world
  • Worldwide distribution eg murine typhus
  • Geographically restricted eg scrub typhus
  • Specific environmental conditions of hosts and vectors
  • Incubation 6 - 14 days (< 21 days)
  • Fever, malaise, headache, myalgia
  • No apparent focus of infection
  • Eschar? Rash? Severe - vasculitic syndrome
  • Variable severity

Bloods
* Normal white cell count
* Normal or low platelets
* CRP elevated
* Normal or slightly elevated ALT/ bilirubin
* Renal failure and DIC late

1518
Q

Key clinical presentation of rickettsial infections?

A

Fever
Rash
Headache

?Lymphadenopathy

+/- Inoculation eschar (if present then
diagnostic)

Incubation 6-10 days (<21d)

1519
Q

Differentials for rash on palms and soles?

A

R.rickettsia
R.conorii
Syphilis
Enterovirus - hand foot and mouth
Mpox
Human monocytic ehrlichiosis
Drug reaction

1520
Q

Tick bite prevention advice?

A

Wear long clothing.
* Check for ticks after outdoor activities.
* DEET
* Promptly/safely remove ticks to prevent transmission of some diseases

If you see a tick
- grab by mouthpart and pull directly upwards, remove mouthpart fully

1521
Q

Tick borne diseases?

A
  • Rickettsial disease eg scrub typhus
  • Babesiosis
  • Borreliosis
  • Crimean Congo Haemorrhagic Fever
  • Lyme disease
  • Tick-borne
    encephalitis
  • Tularaemia
1522
Q

Vector of Rickettsia rickettsii?

A

Ticks (Dermacentor variablis/andersoni, Rhipicephalus)

1523
Q

Vector of Rickettsia conori
(Mediterranean fever, Boutonneuse fever)?

A

Tick
Rhipicephalus sanguineus

1524
Q

Acronym for causes of Loeffler’s syndrome

A

S: strongy
A: ascaris (most common)
N: ecator
T: oxcara canis
A: nyclostoma

1525
Q

Can you use anti-Leish Abs to screen for visceral leishmaniasis?

A

No
a significant proportion of healthy individuals will have anti-Leishmania
antibodies, either from having past treatment for visceral leishmaniasis or having had subclinical infection, so the presence of antibodies is not diagnostic of active disease

1526
Q

What happens after measles infection to the immune system?

A

Profound and prolonged loss of immunological memory

1527
Q

What vaccine coverage is needed for measles elimination?

A

WHO considers that to maintain the interruption of measles virus circulation, immunization programs must provide measles vaccine to at least 95% of each new birth cohort of infants before the age of 2 years

1528
Q

Rickettsia rickettsii
- Distribution
- name of disease
- vector

A

USA

Rocky Mountain
Spotted Fever

Tick- Dermacentor
variablis
American dog tick

1529
Q

Incubation period for plague

A

1-6 days

1530
Q

Rickettsia africae
- Distribution
- name of disease
- vector

A

Africa/
Caribbean
African tick bite fever

Tick- Ambylomma

1531
Q

Rickettsia conori
- Distribution
- name of disease
- vector

A

Worldwide
Europe, Africa, Asia

Mediterranean spotted fever
Boutonneuse fever

Tick Rhipicephalus
sanuineus

1532
Q

Rickettsia akari
- Distribution
- name of disease
- vector

A

Worldwide

Rickettsia pox

Mite- Liponyssoides
sanguinues

1533
Q

Rickettsia typhi
- Distribution
- name of disease
- vector

A

Worldwide

Murine typhus

Flea faeces- Xenopsylla
cehopis

1534
Q

Rickettsa prowazekii
- Distribution
- name of disease
- vector

A

Worldwide

Epidemic typhus

Louse faeces- Pediculus
humanus corporis

1535
Q

Scrub typhus
- cause
- distribution
- vector

A

Orientia tsutsugamushi

Worldwide

Trombiculid Mite

1536
Q

Infection linked to bats and patient getting encephalitis and change in mental state in South Asia?

A

Nipah virus

1537
Q

Swimmer’s itch is due to skin penetration by

A

infective stage cercariae (Schisto)

1538
Q

Microscopy of a warm stool sample is useful in the diagnosis
of

A

amoebiasis

1539
Q

How can you tell ecological studies apart from other study designs?

A

unit of analysis in this case is at the level of the population/community or group as opposed to the other study designs, for example cross- sectional where individuals are the unit of analysis

1540
Q

Define sensitivity

A

the probability of testing positive if diseased

1541
Q

Define specificity

A

the probability of testing negative if not diseased

1542
Q

Define negative predictive value

A

The probably of not being diseased if the test is negative

1543
Q

Clinical features Loiasis

A
  • Typical: migratory loiasis
    ○ Eye worm migration (1/2 day to 3/4 days)
    ○ Calabar swelling (dermal migration of adult worm) - transient, erythematous, non-pitting oedema
  • Common but unspecific
    ○ Asthenia and fatigue
    ○ Severe headaches, transient nerve palsies
    ○ Arthralgia, myalgia, transient toothpain
    ○ Pruritis and rash
  • Rare but important complications
    ○ Vision loss
    ○ Encephalitis
    ○ Nephropathy
    ○ Cardiomyopathy
    ○ Pulmonary inflammation
    ○ Septic arthritis
    ○ Splenic granulomatous lesions
  • Varying clinical manifestation
    Asymptomatic, symptomatic, complicated
1544
Q

Life cycle Loa Loa

A

Bite of crysops fly
Infective L3 larvae develop into adult worms in soft tissue
Migration of adult worms around body (lifespan 20 years)
Mating and offspring produced -> microfilariae which circulate in blood
Crysops take blood meal
L1, L2 in thoracic muscles
L3 in head and mouthpart

1545
Q

How does encephalitis present with Loa Loa and what are the causes?

A

Spontaneous encephalitis
Treatment associated encephalitis

  • Associated with high microfilarial load
  • DEC
  • Ivermectin
  • (Albendazole)

Signs&symptoms:
* Palpebral haemorrhages, CRP
* Fever, headache, confusion, agitation, coma

1546
Q

What is the difference in clinical presentation between endemic regions and returning travellers?

A

Returning travellers
* More often symptomatic
* Higher inflammatory response
* Higher psychological distress
(in patients and doctors…)
Less chronic/long term disease

1547
Q

How do you diagnose loaisis?

A

Adult worm: clinical observation, ?eye migration, calabar swelling (more helpful in travellers)

Mf: blood sample (10am-4pm) microscopy, PCR, LAMP, (LoaScope)
(may be more helpful in endemic areas but NB: only 1/3 will have positive blood for microfilariae)

Serology useful in returning travellers from non-endemic regions but lots of cross reaction due to co-infections in endemic regions

1548
Q

What is the LoaScope and what it used for?

A

Identify patients with Loa Loa as part of Oncho treatment programmes to NOT treat them

1549
Q

What is occult loaisis?

A

Occult: fairly sure they are infected but cannot detect microfilariae in the blood

often misdiagnosed as negative blood tests

1550
Q

What is DEC active against in Loa Loa?

A

Microfilariae (+++) and macrofilariae (++)

1551
Q

What is ivermectin active against in Loa Loa?

A

ONLY microfilariae

1552
Q

What is albendazole active against in Loa Loa?

A

some mild activity against macro and microfilariae

1553
Q

What is doxy active against in Loa Loa?

A

Nothing
Loa Loa have no wolbachia so no role of doxy

1554
Q

Vector of Loa Loa?

A

Deer fly: Chrysops silacea, C. dimidiata
Wood fire, day-biting

1555
Q

Epidemiology of Loa Loa?

A

Forest and adjacent savannah regions of

Central Africa (parts of W-Africa)
~20 million patients, often life-long infection
eg DRC, Gabon

Rural populations

1556
Q

How to treat Loaisis?

A

DEC
Mazzotti reaction, encephalitis in patients with high mf
Contraindication in onchocerciasis
indicated in patients with mf<2,000/ml
3-weeks treatment regimen (60%-70% cure rate)

(Ivermectin)
very rapid and potent activity on mf, no activity on adult worms:
Mazzotti reaction, encephalitis in patients with high mf
indicated in patients with mf <8,000/ml
Single dose; NOT CURATIVE

Albendazole
very slow activity against mf (probably sterilizing effect), low activity against adult worms
Encephalitis? (hasn’t been used that much so poor data)
indicated if DEC (and IVM) cannot be used
3-5 weeks regimens, cure rate?

Tx-algorithm based on microfilaria count; apheresis in hypermicofilaraemia

  • Several cycles to mechanically remove microfilariae
  • Lowering mf below 2,000 (8,000)/ml
  • Consecutive definitive treatment with DEC

Risk-benefit analysis?
In- versus outpatient?

1557
Q

Prevention and prophylaxis for Loa Loa?

A

Avoidance of fly bites (mosquito nets, clothes, repellents)

Very-high-risk travellers: weekly DEC prophylaxis

No control prgrammes in
endemic countries! Also not a NTD.

PERSONAL LEVEL:
Avoidance of deer fly bites
* Clothing, mosquito net cloths
* Repellents?
* Citridiol > DEET > Placebo = Icaridin
Chemoprophylaxis:
* 300mg DEC weekly
* 100% protective efficacy
* Indication:
* selected cases of highest risk
* Peace corps, missionaries, military, forest worker
__
CONTROL
Deforestation and human land-change
* Long-term reduction of transmission regions

Currently available control-tools are insufficient
* MDA-drugs: safety risks and/or not efficacious and/or inappropriate regimens
* Vector: no tools available for efficient vector control

No control programs implemented in endemic regions

Priority for future
* Develop new tools for vector control, mass drug administration programs
* Distribute knowledge about disease (mode of transmission)

1558
Q

What is the HEARTs initiative and what disease is it for?

A

For CVD in primary care

Healthy lifestyle counselling
Evidence based protocols
Access to essential medicines
Risk based CVD management
Team based care
Systems for monitoring

1559
Q

Types of insulin

A

Analogue insulin: rapid-acting (eg. lispro) & long-acting (eg. glargine) -> availability limited in LMICs

Soluble (regular) insulin: short-acting, suitable for SC or IV administration

Isophane (NPH) insulin: intermediate-acting -> cost effective option

Lente insulin: intermediate to long-acting -> used when NPH is unavailable

1560
Q

Issue with SGLT-2 inhibitors?

A

promote glucose excretion but risk euglycemic ketoacidosis in insulin-deficient patients

1561
Q

WHO differentiate service delivery models (DSD)?

A

Individual Models (Facility-based): Patients attend facilities individually for care.

Individual Models (Community-based): Care provided outside facilities, often closer to homes.

Group Models (Healthcare worker-led): Groups of stable patients meet with a health worker for care.

Group Models (Patient-led): Patient groups self-manage their care with provider guidance.

1562
Q

What is the difference between eradication and elimination?

A

Elimination: Reduce incidence of disease to zero in defined geographical area. Continued interventions required to prevent re-introduction

Eradication: Permanent reduction to zero of the worldwide incidence of infection caused by a specific agent

1563
Q

What makes an NTD suitable for elimination?

A

No animal reservoir

Of public health importance

Effective intervention

Amenable to surveillance

1564
Q

Most common features of bancroftian filariasis?

A

Hydrocele
Acute adenolymphangitis
Lymphoedema

1565
Q

What are the different types of poor quality medicines?

Three types of approach to trace the origin of falsified medicines?

A

Falsified: medicines that are deliberately and fraudulently produced

Substandard: Unintentional errors in production or negligence means medicines of poor quality

Degraded: left factory as good quality but deteriorated through not following Good Pharmacy practice in the distribution chain and poor storage

ORIGIN
1. Genomic eg environmental DNA
2. Mass spec
3. Social network analysis

1566
Q

Causes of diffuse unilateral sub retinal neuroretinitis?

A

Gnathostoma (12.5mm)

Baylisascaris procyonsis (1500-2000 micrometres)

Ancyclostoma caninum (500-700 micrometres)

Strongyloides stercoralis (600 micrometers)

Toxocara canis (400micrometres)

1567
Q

Baylisascaris
- What is it?
- Epi?
- Infection syndromes in humans

A

Zoonotic infection from racoon ascaris

Most common: Baylisascaris procyonis

Epi: North America, Europe and Japan

In animals:
- If ingested by other animals then makes them display abnormal behaviour which makes them easier prey for racoons
- Not harmful to the raccoons

In humans:
- Infections rare, only if OLD faeces from racoons ingested (eg >2 weeks
- Often asymptomatic but can cause 3 major syndromes
1. Visceral larva migrans: eosinophilia and organ involvement
2. Neural larva migrans - predilection for neural tissue leading to eosinophilic meningitis
3. Ocular larva migrans - DUSN (diffuse unilateral subretinal neuritis)

1568
Q

Stages of Baylisascaris

A

Stages of disease:
1. Multi-focal grey white lesions
2. Late stage disease: pigmentation alterations, narrowed retinal vessels, pale optic disc
Ideally worm visualisation: 1500-2000 mcm, occurs in approximately 25-40% of cases, size 400-2000mcm

1569
Q

Broader differentials for diffuse unilateral subacute neuroretinitis

A
  • Toxoplasmosis
  • Histoplasmosis
  • Syphilis choroiditis
  • Multifocal choroiditis
  • Serpinginous choroiditis
  • Acute posterior multifocal placoid pigment epitheliopathy
  • Multiple evanescent white dot syndrome
  • Optic neuritis
1570
Q

Treatment of Baylisascaris

A

Treatment
1. Neurological
- Albendazole and praziquantel for prolonged duration +/- steroids
- Outcomes poor
- High risk exposure: albendazole prevention 10-20 days

  1. Ocular (DUSN)
    - Photocoagulation
    If not possible -> albendazole +/- scattered photocoagulation
1571
Q

Life cycle trypanosoma cruzi

A

A. Dividing epimastigotes
B. Infective metacyclics
C. Excreted infective metacyclics
D. Host cell infection
E. Circulating trypomastigotes
F. Pseudocyst containing dividing amastigotes
Ingestion of trypomastigotes by insect vector

1572
Q

How to make a diagnosis of T cruzi?

A
  • ELISA
  • IFAT (indirect immunofluorescence Ab test)
  • PCR
  • RAPID tests (immunochromatographic dipstick test eg STATPACK)
  • Xenodiagnosis

In chronic phase need two accordant serological tests for diagnosis

In essence:
- Acute: PCR or microscopy (rarely positive) or RDT
- Chronic: 2x serological test

1573
Q

What is ocular surface squamous neoplasia and what are its risk factors?

A

Dysplastic lesions of conjunctiva and cornea (ranges from conjunctival intraepithelial neoplasia to invasive SCC of conjunctiva)

RFs: HIV (esp in sub saharan Africa) and UV exposure

Either treat with topical chemotherapy agents to begin with or if invasive needs surgery which may mean removing whole orbit

1574
Q

Epi of fungal infections?

A

Now estimated 3.8 million deaths from fungal infections

1575
Q

4 critical fungi on WHO fungal priority pathogens list?

A
  • Cryptococcus neoformans
  • Candida auris
  • Aspergillus fumigatus
  • Candida albicans
1576
Q

Gold standard for fungal infection diagnosis?

A

Histopathology

1577
Q

What do echinocandins target?

A

Beta-glucan chains

eg Caspofungin

1578
Q

What fungal infections will Beta-D Glucan be negative in?

A

Crytococcus

Mucormycosis

1579
Q

What fungal infections will galactomannan be negative in?

A

Invasive candidiasis

PCP

Cryptococcosis

Mucormycosis

1580
Q

Rapid diagnostic tests currently available for fungal infections?

A
  • CRAG: very sensitive and specific for cryptococcus
  • Histoplasma urinary antigen LFA
  • Aspergillus GM LFA
  • Coccidioides Ab LFA
1581
Q

Does fluconazole work on yeasts or moulds?

A

Only yeasts

1582
Q

What 3 main classes of drugs do we have to treat serious fungal infections?

A

Polyenes
- Nystatin
- Amphotericin B
- Liposomal amphotericin B

Echinocandins
- Caspofungin etc

Azoles
- Fluconazole
- Itraconazole
- Voriconazole etc

Other
- Flucytosine
- Terbinafine

1583
Q

First line treatment globally for skin and nail fungal infections?

A

Terbinafine

1584
Q

Risk factors/associations of mucormycosis?

A

Steroid use

Diabetes

Neutropenia

1585
Q

Diagnostic tests and issues for histoplasmosis?

A

TB mimic
Massively underdiagnosed especially in African countries due to lack of diagnostics

Diagnostic tests:
- Culture (slow and needs containment level 3)
- BD Glucan and galactomannan will be positive
- Serology -> less useful in HIV/disseminated diseases
Ag testing in blood and urine (future but not widely available even in rich countries)

1586
Q

Treatment of histoplasmosis?

A

Disseminated: Ambisome/amphotericin

Localised, pulmonary: Itraconazole

1587
Q

Clinical manifestation of Blastomycosis?

A

Blastomycosis
- Pneumonia most common manifestation (60-90%)
- Extra-pulmonary manifestations common
○ Myalgia, arthralgia, chills
- Dissemination to skin also reported frequently

NB: Ontario in Canada world hotspot

1588
Q

Where is a world hotspot for blastomycosis infections?

A

Ontario in Canada

1589
Q

Basic functions of the skin

A
  • Barrier
  • Immune function
  • Temperature regulation
  • Attraction/display
  • Sensing the environment
  • Metabolic
  • Homeostasis of water, electrolytes
1590
Q

Common skin disorders (defined by International foundation for Dermatology and the WHO)

A
  1. Scabies
  2. Bacterial skin infection
  3. Fungal infection (superficial infection)
  4. Eczema
  5. Acne
  6. HIV-associated skin diseases
  7. Pigmentary disorders
  8. Wounds
1591
Q

Scabies
- Causative agent
- Epi
- Transmission

A

Causative Agent

Sarcoptes scabiei var. hominis
Ectoparasitic mite

WHO NTD in 2017

Epi

565 million people
4.84 million DALYs

Transmission
Direct (most common): close physical contact

Indirect:
- fomites and bedding
- outbreak in closed communities

1592
Q

Clinical features of scabies

A

Clinical Features

Symptoms: thought to be due to hypersensitivity to mite, eggs, faeces
- Ie continual exposure of mites burrowing into skin causes sensitisation and resulting allergy

Intense itching, often worse at night
Burrows* (thin, greyish-white, raised lines), typically seen in skin folds

Papules, nodules, vesicles, pustules

Excoriations

Pattern may differ in children and elderly
- eg soles of feet in infants

Secondary bacterial infections can occur due to scratching

1593
Q

Two different types of scabies?

A
  1. Typical Scabies: Localized skin infestation
  2. Crusted Scabies (Norwegian Scabies): Severe, highly contagious form with thick crusts, common in immunocompromised patients eg HIV, HTLV-1
  • Hyperkeratosis
  • Itch may NOT be prominent feature
  • (esp in elderly and children)
1594
Q

Diagnosis and treatment of scabies?

A

Clinical: rash and burrows
Dermatoscope: delta wing sign
Microscopy: mite or eggs

No serological, molecular, POC tests
Peripheral eosinophilia

TREATMENT
Practical steps in treating scabies
* Information
* Diagnosis
* Transmission
* Fomites

Treatment advice
* Benzyl benzoate 3 consecutive days concentration determined by age (most available in LMICs)

OR

TWO treatments 7 days apart of either
* Permethrin 5% (esp babies)
OR
* Ivermectin 200µg/kg

Apply creams from neck down (head to toe for infants and elderly)
Household contacts treated simultaneously
Expectations from treatment and management of post-treatment symptoms (itching won’t go away for at least 6 weeks) => post scabies itch and dermatitis
Provide psychological support and sexual health screen (to adolescents and adults, including HIV test)

For crusted scabies:
- Oral: Ivermectin (dosing depending on grading)
Topical: permethrin or benzyl benzoate (long course eg weeks) + keratolytic eg urea/lactic acid or salicyclic acid

1595
Q

When is MDA appropriate for scabies and what do you treat with?

A

WHO informal consultation 2019
* community prevalence of scabies is ≥ 10%
* Two doses of oral ivermectin delivered 7–14 days
* 3–5 rounds of MDA
* Stop if prevalence of scabies falls < 2%

1596
Q

Complications of scabies

A

Secondary bacterial infections (e.g., impetigo, cellulitis)
Post-streptococcal glomerulonephritis
Bullous disease
Erythroderma
Widespread skin loss

1597
Q

What is the difference between visualising fungi in culture or in tissue?

A

Both you are visualising fungi in asexual state

In tissue: visualise fungi as hyphae and yeasts

In culture: visualise as spores

1598
Q

What causes Tinea infections and give some examples of types? Which two types have equal split of causes between human and zoonotic causes?

A

Caused by keratophilic moulds

Most common causative fungus: Trychophyton rubrum

Examples:
*Tinea capitis
*Tinea faciei
Tinea corporis
Tinea pedis

  • roughly 50% caused by non-anthropophilic dermatophytes
1599
Q

What is the difference between endothrix and ectothrix?

A

Endothrix: hyphae are within the hair
Eg Trichophyton sp.

Ectothrix: hyphae outside of the hair
Eg Microsporum sp

1600
Q

What is a dermatophyte species that is associated with widespread resistance?

A

Trichophyton indotineae

1601
Q

What does Hortaea werneckii cause?

A

Tinea nigra
Saprophyte - found in soil, wood, compost

Asymptomatic infection of the skin (palms>soles)
Rare, adults>children

Tx: topical azole eg ketoconazole

1602
Q

Two different causes of mycetoma? And areas of endemicity?

A

Eumycetoma = fungal
- in Africa

Actinomycetoma
- in Americas

1603
Q

Commonest cause of eumycetoma?
Characteristics of infection?

A

Madurella mycetomatis

  • DARK grains
  • Slow
  • Walled off
  • Late invasion
  • Punched out lesions on XR
1604
Q

Treatment of madurella mycetomatis?

A

Itraconazole (400mg OD) for years + surgery
Recurrence in >1/3

Trial posconazole in refractory disease

1605
Q

Treatment of actinomycetoma?

A

Co-trimoxazole

Can use in combo with
- amikacin
- rifampicin
- co-amoxiclav

1606
Q

Characteristics of Viriperidae

A

rough scales
slit like eyes
large head
short, thick, short-tailed
colourful dorsal pattern - like warning signs

slow moving but lightening striking

1607
Q

Biggest bitter in East Africa

A

Puff adders (have repeating V or U pattern on back)
Grumpy and won’t move but will bite and sting fast

1608
Q

Characteristics of elapidae

A

Bands of colouring
No warning signs
Can have hoods

1609
Q

Key toxic snake in South East Asia

A

Kraits *part of elapidae

1610
Q

Diagnosis of mycetoma

A

Biopsy and histopathology
or
PCR

1611
Q

Leading bacterial cause of mycetoma and characteristics

Tx?

A

Nocardia (asterioides)
WHITE grains

Mostly feet, trunk and head possible
Rapid and spreading infection with unclear margins, inflammation and destruction

Rapid spread to bone so hard to treat

Mx:
- Co-trimoxazole
- Often add amikacin or rifampicin

1612
Q

What is myiasis caused by in South America?

A

Bot fly
Dermatobia hominis

Often need to incise as too big to do just vaseline

1613
Q

Polio
- Causative agent?
- Transmission?
- Pathophysiology

A

Causative Agent

Poliovirus = enterovirus, RNA virus
Three serotypes: PV1 (most virulent), PV2 (eradicated), and PV3
1 most virulent > 3 > 2

Transmission

Fecal-oral route (primary) or via respiratory droplets
Facilitated by poor hygiene and sanitation
Case infection ratio low (1/100 to 1/2000) and increases with age + more likely to be paralysed

Pathophysiology

Virus to mucosa
Then to lymphoid tissue then CNS -> acute flaccid paralysis (AFP)

1614
Q

Clinical features of Polio

A
  1. Asymptomatic (90-95%)
  2. Minor illness: Fever, malaise, sore throat, headache, and vomiting
  3. Non-paralytic poliomyelitis: Aseptic meningitis with neck stiffness, back pain, and muscle spasms
  4. Paralytic poliomyelitis: Flaccid paralysis due to anterior horn cell damage; affects limbs asymmetrically; can involve respiratory muscles
  5. Post-polio syndrome (PPS): Late-onset muscle weakness and fatigue years after recovery
1615
Q

What was the eradication strategy for Polio?

A

as part of Millennium development goals - to eradicate Polio

  1. High routine immunisation coverage
    emphasis on OPV
  2. National Immunisation Days (NIDs)
    OPV to all children under five
    more than 1 billion doses of OPV per year….
  3. Acute Flaccid Paralysis (AFP) surveillance
    expect at least 1 per 100,000 under age 15
  4. » Mop-up » campaigns
    house to house
1616
Q

Differences between two Polio vaccines

A

Oral Polio Vaccine (OPV): Live attenuated vaccine providing intestinal immunity; used in eradication campaigns
TRIVALENT to BIVALENT since 2016
- Less expensive, virus builds up in gut to increase antigenic mass = greater enteric immunity (vs infection)
- Less expensive than IPV
- Transmissible: so immunises > vaccinated (ie virus comes out of faeces as still live in kids who vaccinated so can then be spread and vaccinate whole household but turned out not to be true ie creation of vaccine derived polio vaccines)

BUT
- Lower seroconversion in tropics ?due to competing organisms in the gut
- Causes paralysis approx 1 per million doses
- Transmissibility is two edged sword
Another issue
- OPV is good at stopping clinical disease but not necessarily transmission so when OPV campaigns stopped after coverage given, Type 1 poliovirus found in sewers etc (Israel) suggesting asymptomatic carriage

Inactivated Polio Vaccine (IPV): Injected vaccine providing systemic immunity; used in routine immunization schedules

1617
Q

Diagnosis of Polio?

A

Diagnosis

Clinical
Laboratory confirmation via virus isolation from stool, throat swabs, or CSF
RT-PCR for viral RNA

(Serology does not differentiate between old and new/immunised)

Treatment

No antiviral treatment available
Supportive care: Pain relief, physical therapy, and respiratory support if needed
Prevent secondary complications like contractures and infections

1618
Q

What are vaccine derived polio virus?

A

Polioviruses that result from vaccination (esp in areas of low immunisation rates) which take on wild type characterictis and complicated elimination strategy

1619
Q

Current situation with eradication programme of Polio and key endgame challenges?

A

Wild type circulating in 2 countries: Pakistan and Afganistan
VDPV circulating in 20 countries

Endgame challenges:
1. To stop all wild virus circulation
2. To stop all VDPV circulation via NOPV2 (novel oral polio virus vaccine 2 since 2021)
3. Security against reintroduction
4. To stop all vaccination

1620
Q

Complications of Poliovirus infection?

A

Paralysis, which can be permanent

Bulbar poliomyelitis leading to respiratory failure

Post-polio syndrome with late-onset weakness and fatigue

1621
Q

Recommended vaccine schedule for Polio virus

A

3 doses
(can give additional birth dose can be referred to as 0, so gives 4 doses)

  • can have in addition to routine OPV dose to kids up to 5 years old
1622
Q

What does appropriate footwear eg fang proof boots protect against?

A

Snake bites
Hookworm
Strongyloides
Podoconiasis
Tungiasis
Venomous bites and stings

1623
Q

What is Tungiasis caused by?

A

Tunga penetrans
Jigger flea –> adults burrow into skin via soil

1624
Q

How to prevent against Tungiasis?

A

○ Treat patients
○ Sealing/spraying floors with insecticide
○ Encourage footwear
○ Treat infected animals owned by infected families

1625
Q

How does sporotrichosis present?

A

75% lymphocuteanous
Ulcerating nodules, plaques on exposed body sites eg arms and legs

25% fixed cutaneous
variable morphology of skin lesions eg papulopustular, nodular, verrucous

Can also be non-cutanaeous with localised/systemic form eg ocular, tenosynovitis, CNS

1626
Q

Pathogenic causes of Sporotrichosis?

A

Sporothrix schenkii
Sporothrix braziliensis (from cat scratch eg in Brazil)
Sporothrix globosa

1627
Q

Treatment of sporotrichosis?

A

Most commonly: itraconazole (at least 3 months)
or terbinafine
or potassium iodide

Treat until all lesions resolve

Consider:
Cryotherapy
Thermotherapy
Photodynamic therapy

1628
Q

Who is at risk of chromoblastomycosis?

A

Adult males
Agricultural workers
Immunocompetent

High in Brazil, China, Mexico

1629
Q

Where is Lobiasis found, what causes it and how is it treated?

A

Lobomycosis – Lacazia loboi
Central and South America

Treat with excision of lesion

1630
Q

What are the two main types of Entomophthoromycosis?

A

Slow progressing
Disfiguring
Inoculation or inhalation
Sub cut swelling or fibrosis

Basidiobolus ranarum
- SE Asia, Africa
-Limb swelling
- Children eg boys

Conidiobolus coronatus
- Africa, India, West Indies, South America
- Facial swelling
- Young adults (male)

1631
Q

What is African Histoplasmosis caused by and how does it present?

A

Histoplasma duboisii
Intracellular parasite, yeast
Transmission: inhalation

Presentation: bone, LN, skin lesions
Do X-rays to look for bone lesions

1632
Q

What is blastomycosis caused by? How is it transmitted?
Who is at risk?
How is it present?

A

Blastomyces dermatitis
Blastomyces gilchristii

IN soil and organic matter -> INHALATION

Agricultural workers
Men>women
Uncommon in HIV/AIDS

PRESENTATION
1) Primary cutaneous -> very rare
2) Pulmonary blastomycosis (eg cavitating lesion, abscess)
3) Disseminated eg widespread skin rash

1633
Q

Investigations for Blastomycosis?

A

Microscopy -> thick-walled, spherical yeasts

Culture -> mould at 30degrees, white tan cotton appearance

1634
Q

Diagnosis and treatment of coccidioidomycosis?

A

Microsopy -> Large sphericals containing endospores

Culture -> fast growing white-tan cotton appearance OR thick-walled arthroconidia

Treatment
- Pulmonary: not always necessary
Otherwise:
- Itraconazole or fluconazole
- Ambisome/Amphotericin
Disseminated: complement fixing Abs

1635
Q

Where are 80% of the world’s paracoccidioidomycoses cases found?

A

Brazil

1636
Q

What are the two clinical forms of paracoccidioidomycosis?

A

Juvenile form of paracoccidioidomycosis
- Lymphadenopathy
- Fever
- Skin lesions in 50%
- Hepatosplenomegaly

Adult form
Vegetative lesions
- Lesions in oral mucosa
Macrochelitis
- Lesions in nose/eyes
- Skin lesions across body

1637
Q

Investigations and treatment or paracoccidioidomycosis?

A

INVESTIGATIONS
Direct visualisation via microscopy: Mickey mouse appearance or sailor ship

Culture

TREATMENT
- itraconazole
- ambisome
- voriconazole
- co-trimoxazole

1638
Q

Treatment seborrheic dermatitis?

A

Clotrimazole 1%
Hydrocortisone

Other
- Econazole cream
- Ketoconazole cream/shampoo

1639
Q

Treatment of eczema (WHO essential drugs)

A

Betamethasone valerate
Hydrocortisone

1640
Q

WHO essential medicine for PPE in HIV?

A

Betamethasone

Papular pruritic eruption
- associated with low CD4 and high VL

1641
Q

Different types of Kaposi’s sarcoma

A
  • Classical (old people)
  • Endemic (was before HIV/AIDs)
  • HIV/AIDS
  • Transplant
1642
Q

WHO essential medicines for Kaposi’s sarcoma

A

ART (may be enough alone if not severe disease)
Doxirubicin/Adriamycin
Bleomycin
Vincristine
ABV

1643
Q

WHO essential medicines for warts?

A

Salicylic acid
Podophyllum

1644
Q

WHO essential medicines for psoriasis?

A

Betamethasone and ART

1645
Q

WHO essential medicines for eosinophilic folliculitis?

A

Think very low CD4 counts who have started therapy

Itraconazole *despite not being fungal disease
Permethrin

1646
Q

What CD4 counts do you get eosinophilic folliculitis?

A

<200

Patients get within starting ART ie within 3-6 months

VERY ITCHY

1647
Q

How can Herpes simplex present in people living with HIV?

A

Ulcerated lesions with scalloped borders
- Transmission and acquisition in PLWH 4x more likely
- Can also genital/perioral transmission

Treat with acyclovir

NB: aciclovir resistant herpes is 10x more common in immunocompromised -> can present as SCC mimicking

1648
Q

Who gets molluscum with HIV?

A

Low CD4 count
May be extensive
Often facial involvement

1649
Q

Skin lesions identified in Mpox?

A

POLYMORPHIC ERUPTION
Pustules
Pseudopustules
Ulcers

1650
Q

How might prematurity influence CKD burden?

A

Prematurity can reduce nephron endowment -> higher risk of CKD

Thus, risk of burden in LMICS

1651
Q

At what point does acute kidney disease (AKD) be classified as CKD?

A

If kidney injury for >90 days

1652
Q

Definition of CKD

A

Abnormalities of kidney structure or function present for >3 months with decreased eGFR (eGFR<60) AND/OR markers of kidney damage (one or more)

1653
Q

Signs and symptoms of CKD?

A

HTN
Pruritis
Nocturia
Dyspnoea
Lethargy
Vomiting
Body swelling
Anorexia
Restles legs
Haematuria

1654
Q

What measure should you use to assess albuminuria?

A

Urine creatinine to albumin ratio

1655
Q

What are the two molecules used to calculate eGFR?

A

Creatinine OR Cystatin C (better in SSA)

1656
Q

Causes of AKI (esp in LMICs)

A
  • Sepsis (very common in LMICs) -> Malaria, TB, Bacterial etc
  • Toxins (nephrotoxins)
  • Obstruction
  • Parenchymal kidney disease
1657
Q

Two key examples of lice and relevant risk factors?

A

Pediculus (head and clothing/body lice)
Pthirus (crab lice)

*Pediculus humanus (body lice) - uncommon in developed countries (usually vagrants)
*Pediculus capitis (head lice) - world-wide
(mostly in children)
*Pthirus pubis (crab lice) - mainly in sexually
active groups, can occur on beards and eye
lashes

1658
Q

Life cycle of lice and where they are to be found?

A

LIFE CYCLE
Egg - dark (empty egg =nit is white), laid at scalp where its warmest, nymph (3 instars), adult (9 days longevity)
Obligate blood feeders; feed multiple times daily

FOUND
- Clothing (hems and seams)
- Search hair on bed (wet combing with conditioner)
- Search coarse hair (pubic, beards and eyelashes/eyebrows)

1659
Q

Medical importance of lice?

A

P.humanus
- Transmit louse-borne relapsing fever, agent is spirochaete Borrelia recurrentis)
- Rickettsia prowazekii (typhus): multiples in midgut, passed in faeces (infected for 3 months). Anthroponotic typhus.
- Trench fever (Bartonella quintana)

Pediculosis = infestation with louse
- Pediculosis capitis = head lice, heavy infestations cause irritation, itching & saliva toxic reaction
- Pediculosis pubis -> characteristic bluish spots at bite site. Associated with STIs.

1660
Q

Control of lice - methods for
- P humanus
- P capitis
- Pthirus

A

P.humanus
Environmental
- Replace clothes/isolate for 17+ days
- Boil wash clothes
- Hot tumble dry (60 degrees for 15mins)
Chemical
- Insecticide treatment of clothes
- If in prision: cleanse prison, shave and dust prisoners with 0.5% permethrin, change beds and clothes

P.capitis
Chemical
- Insecticide resistance to all over-the-counter treatments containing either malathion, permethrin or phenothrin is widespread and ovicidal activity is poor
- Dimeticone 4% gel/lotion – prevents respiration (no resistance)
Physical
- Using wet-combing with conditioner is more effective
* Treat all individuals in household with an active infestation.
* No need to treat clothing/bedding.

Pthirus
Eyelashes
- Petroleum jelly 2xday for 10 days
Pubic hair and beards
Water-based insecticide lotions (alcohol-based lotions sting delicate areas)

1661
Q

Relevant families of flea and how to tell them apart?
How to tell female vs male apart?

A

How to tell them apart?
- Difference in pronotal and gental combs and mesopleural rods
- Female: round abdomen, spermathacea
- Male: Terminal segments of male abdomen
are upturned, Aedeagus – intromittent organ

Types:

  • Xenopsylla (plague flea)
    ○ No combs
    ○ Mesopleural rod in second thoracic segment
  • Pulex irritans (human flea)
    ○ No combs
    ○ Sclerotised antennal fossae (like wearing headphones)
  • Ctenocephalides (cat/dog flea)
    ○ Genal and pronotal comb
    ○ Mesopleural rod
    ○ Antennal fossae are not sclerotized
  • Tunga penetrans (Jigger flea)
    ○ Rounder body shape
    ○ No combs
    ○ Compressed thoracic segments
    ○ Head has upturned point
    ○ Mouthparts are long and serrated
1662
Q

Life cycle of fleas and behaviour

A

Egg, larva, pupa, adult
▪Adult longevity 6-12 months (up to 2 years)

▪Female produces 3-25 eggs/day (300-1000/lifetime)

▪Eggs hatch in 2-14 days.

▪Caterpillar-like larvae undergo 2 moults (3 instars) in 2-3 weeks

▪Pupae housed in silk cocoon (2-3 weeks).

▪Adult sheds pupal skin & remains dormant within cocoon (up to one year) until stimulated by a host (vibration).

▪Can copulate immediately after emergence.

▪Eggs laid 1-2 days after bloodmeal

BEHAVIOUR
Males and females opportunistic blood feeders but can survive for long time without feed
Not host specific (but nest/habitat specific)
Primary host required to achieve sexual maturity and reproduction
Saliva (anticoagulant) injected when mouthparts penetrate

1663
Q

Medical importance of fleas

A
  • Yersinia pestis (plague) via Xenopsylla
  • Rickettsia typhi (murine typhus) -> Xenopsylla but dried faeces causes disease
  • Tungiasis via Tunga penetrans
  • Tapeworms eg Dipylidium caninum, Hymenolepis diminuta via Ctenocephalides spp. (cat/dog flea) -> flea ingested -> give Endogard for dogs (Praziquantel, Pyrantetel, Emonate, Febantel) every 2 weeks until 12 weeks, then every 3 months
1664
Q

Control/prevention of fleas

A

Chemical:
- Domestic fleas: insectide tx of houses
- Pets (treatment) and other hosts
- Reapplication of treatments as reinfestations may appear quickly

Environmental:
- vacuuming, around edges of carpets, washing floors

PPE: shoes, DEET

Rodent control + proofing of buildings

Mini-smoke generator with insecticide ie permethrin 13.5%

1665
Q

Life cycle of bed bugs and behaviour

A

~3 weeks

  • 1st instar nymph hatches from an egg and then goes through 4 further nymphal instars, before moulting to become an adult.
  • Without restrictions due to temperature or host availability, the bed bug life cycle can be completed in 3 weeks.
  • Peak infestation occurs in around September followed by a decline during the winter and a
    steady increase throughout the spring and summer months

BEHAVIOUR
Nocturnal feeders
Obligate blood feeders attracted to warmth and CO₂
During day: aggregate within refuges in conspecific groups consisting of all developmental stages and both sexes

Check sheets/cabinets etc for faecal spots

1666
Q

Medical importance and control of bed bugs

A

IMPORTANCE
- Biting nuisance
- Allergic reactions and secondary infections
- Anxiety
- Economic loss
- Potential vector for transmission of trench fever

CONTROL
* important public health problem
* infestations are increasing
* pesticide resistance is developing
* no passive monitoring device
* new approaches are needed
– control
– monitoring
* semiochemicals for odour-baited traps

1667
Q

Life cycle of
- Dermatophygoides
- Scabies sarcoptei
- Thrombuculid mites

A

DERMATOPHYGOIDES
Egg, hexopod larva, 2/3 octopod nympha; stages (protonymph, deutonymph, tritonymph), adult

SCABIES
Adult lifespan 1-2months
Females lay 1-3 eggs per day
Eggs hatch in 3-5 days
Larvae crawl to surface along burrows
Make moulting pocket in skin or hair follicles
After 3-4 fays larva moult to protonymph and after another 2-3 days to tritonymph
Tritonymph moults to either male or female
Male seeks out females
Females stay put and only burrow after fertilisation

THROMBUCULID MITES

-“8-shaped” nymphs &
adults live in soil (feed on
arthropods & eggs)
-Insemination: walk
over spermatophores
-Eggs laid in damp soil
-Larvae (“chiggers”) fall
on host and feed on cell
tissue fluids through a
stylosome

1668
Q

Four key mites of public health importance?

A

Dermatophagoides (House dust mite: triggers asthma)

Trombiculid mites “chiggers”/harvest mites (transmit scrub typhus)

Sarcoptes scabei (causes scabies)

Demodex (follicle mite)

1669
Q

Control measures for
- Dermatophygoides
- Scabies sarcoptei
- Thrombuculid mites

A

DERMATOPHYGOIDES
- Vaccum sampling to assess densities per unit weight
- Mitest collector
- Simple mite extraction method
- Rapid test for mite allergen
- ELISA to quantify Ags
Control with
Physical
- Semi-permeable mattress encasings
- Removal of domestic habitats/dust eg vaccums, ionisers
- Making habitats less favourable for HDM eg de-humidifiers, removing soft furnishings
Chemical
- Allergen sprays
- Fungicides
- Acaricides

SCABIES
- Treat with permethrin 5% cream or benzyl benzoate (whole body below neck, before bedtime, repeat after 7 days)
- MDA: If prevalence is >2%, 2 doses of oral ivermectin (200 μg/kg)
given 7 to 14 days apart

THROMBICULID MITES
- Avoid Chigger habitats/mite islands (patchy distribution: need environment with abundant prey, for adults, and with mammals, for larvae)
- Spray vegetation with an acaricide (e.g. benzyl benzoate)
- Impregnate socks/trousers with repellent (e.g. Diethyltoluamide = DEET).
Treatment for scrub typhus: doxycycline (2.2 mg/kg/dose bid PO or IV, maximum 200 mg/day for 7-15 days)

1670
Q

Medically important mites and their presentation

A

Dermatophagoides
- Faecal pellets can trigger asthma, eczema, perennial rhinitis

Sarcoptes scabiei
- Itchy, red rash
- Especially where skin is thinning eg folds
- Diagnose with skin scrapes
- Crusting in immunocompromised
- Treat with permethrin 5% cream or benzyl benzoate (whole body below neck, before bedtime, repeat after 7 days)
- MDA: If prevalence is >2%, 2 doses of oral ivermectin (200 μg/kg)
given 7 to 14 days apart

Thrombiculid mites
1. Trombidiosis ( or Scrub itch) 20 spp. of chiggers (or harvest mites) can
cause intense, itchy dermatitis.
- Larvae feed for up to 10 days on host (saliva produces itchy, red lumps found, typically in armpits, leg calves, and pelvic region).
2. Scrub Typhus (or Chigger-borne rickettsiosis)
- Orientia tsutsugamushi
- Typically confined to rodents/insectivores
- Humans typically get infected when they invade “mite islands
- Endemic in SE Asia & Australia.
- Symptoms: mild fever to fatal multiple-organ failure (1 million cases/year, up to 40 mortality)

1671
Q

Two types of tick and control methods

A

Hard ticks: Ixodidae
Soft ticks: Argasidae

Control
* Avoidance.
* Repellents (products with 20%+ DEET).
* Clothing (and permethrin impregnated clothing).
* Check yourself and pets for ticks.
* Safe removal of attached ticks.
* Tumble dry clothing on high heat to kill ticks.
* Insecticidal treatment of animals.
* Insecticidal treatment of breeding sites.

Removal
- Fine forceps
- Grasp close to skin
Pull up directly and don’t twist -> want to pull with head

1672
Q

Medical relevance of ticks?

A

IXODIDAE
Arboviruses
- Tick-borne encephalitis
- CCHF
- Colorado tick fever
Rickettsiae
- Rickettsia rickettsii (Rocky Mountain spotted fever)
- Boutonneuse fever (tick typhus)
- Queensland tick typhus
Spirochaetes
- Borrelia burgdorferi (Lyme disease)

ARGASIDS
* Tick-borne relapsing fever.
* Several Borrelia species
Ornithodoros species
* Man is the sole reservoir of disease (B. duttonii).
* Endemic in Africa and southern Asia.
* Foci in North America and Europe.

1673
Q

Life cycle
- Hard ticks
- Soft ticks

A

HARD
Female feeds 6-12 days
Engorged female drops off host and matures eggs
Female lays 1000-10,000 eggs then dies
Eggs hatch in 10-20 days or may overwinter
6 legged larvae feeds for 2-6 days
8 legged nymph feeds for 3-8 days
Nymph moult into males or females
Males feed only briefly
Lifecycle 2-3 years for most temperate species

SOFT
Little sexual dimorphism
Females lay several small egg batches 15-100 eggs after each bloodmeal
Eggs hatch after 1-3 weeks
1 larval stage
Multiple nymphal stages (4-5) each requires bloodmeal
Adults can survive for 12 years without bloodmeal
Lifecycle: 6-12 months to complete
Lifespan 15-20 years

1674
Q

Diagnostic test for trypanosomiasis

A

Blood films - thick and thin

CSF

1675
Q

Life cycle T cruzi

A

An infected triatomine insect vector takes a blood meal from a mammalian host and releases trypomastigotes in its feces. Trypomastigotes enter the host through the wound or through mucosal membranes. Inside the host, trypomastigotes invade cells , where they differentiate into intracellular amastigotes. The amastigotes multiply, differentiate into trypomastigotes, and are released into the circulation as bloodstream trypomastigotes. Trypomastigotes infect cells and transform into intracellular amastigotes. The clinical manifestations of acute infection can result from this infective cycle. Replication resumes only when the parasites enter another cell or are ingested by another vector. The triatomine bug becomes infected by feeding on human or animal blood containing circulating parasites. The ingested trypomastigotes transform into epimastigotes. The parasites multiply in the midgut and differentiate into infective metacyclic trypomastigotes in the hindgut. Trypanosoma cruzi can also be transmitted through blood transfusions, organ transplantation, transplacentally, and in laboratory accidents.

1676
Q

Mechanisms of AMR?

What is the difference between intrinsic and acquired resistance?

A
  • Prevent drug reaching its target via
  • Impermeable outer membrane / wall
  • Efflux pumps
  • Destroy or modify the compound
  • Alter the target or its product so that the drug has no effect
  • Intrinsic resistance
    e.g. lacks antibiotic target
  • Acquired resistance
  • Point mutations
  • Altered target
  • Random errors during replication, selected by antimicrobial pressure
1677
Q

How can antimicrobial resistance be transmitted?

A

Transmit the organism

OR

Transmit the resistance gene
* Plasmid (circles of DNA that can move between cells)
* Bacteriophage (viruses that attack germs and carry DNA from germ to germ)
* Other mobile genetic elements e.g.
transposons (small pieces of DNA that can move from chromosomes), gene cassettes

Mobile genetic elements work via
- Transduction (transfer via phages)
- Conjugation (via connection of bacteria)
- Transformation (picking up resistance genes from nearby germs)

1678
Q

Key bacteria that are WHO concerns for resistance?

A

S.pneumoniae

S.aureus

E.coli

Klebsiella pneumonia

Salmonella spp

Actinetobacter spp

Shigella spp

N.gonorrhoeae

Mycobacterium tuberculosis

1679
Q

Where is fluoroquinolone resistance rising?

A

Nigeria

1680
Q

Approaches to tackle AMR

A

Reduce use

Reduce disease
- vaccination
- better treatment
- better health and sanitation

1681
Q

Why are LMICs at higher risk/suffer more from AMR?

A
  • Poor sanitation – increased risk of illness and increased risk of AMR
    transmission
  • More adverse consequences when infections do occur
  • Limited access to 2nd / 3rd line treatment
  • Limited access to supportive care
  • Poor infection prevention / control practices increasing dissemination within healthcare
    settings
  • And compounded by lack of diagnostics leading to increased use of available
    agents

Same issues in agricultural illness

1682
Q

Contributing factors to AMR?

A
  • Animal production eg lifestock, aquaculuture
  • Animal health eg pets and lifestock
  • Human health eg disease and use of Abx
  • Food chain
  • Agriculture
  • Travel
  • MDA
  • Suboptimal rapid diagnostics
  • Suboptimal vaccination
  • Environmental contamination
1683
Q

How can we measure AMR?

A
  • Rates of resistant infection eg Hospital and laboratory data
  • Process indicators eg Antimicrobial use via Point prevalence surveys
  • Antimicrobial consumption eg Pharmacy records, Import data
  • Rates of resistance in carriage
    samples E.g. stool screening, MRSA screening
  • Proxy markers of resistance eg Environmental sampling
1684
Q

What are the different sickle cell genotypes and their relevant phenotypes?

A

SCD
- Hb SS -> most severe form
- Hb Sβ0 thalassemia -> Most prevalent in the eastern Mediterranean region and India (also includes severe β+ with <5%HbA present)
- Hb SDpunjab -> Predominant in northern India but occurs worldwide

Moderate SCD
- Hb SC -> 25-30% of cases of SCD in populations of African origin
- Hb Sβ+ thalassaemia -> Most cases found in the eastern Mediterranean region (6-15% HbA present)

Mild SCD
- Hb Sβ++ thalassaemia -> Mostly in populations in Africa (16-30% HbA present)

Very mild SCD
- Hb S/HPFH* Group of disorders caused by large deletions of the β-globin gene. Typically, >30% HbF

1685
Q

Diagnosis of sickle cell disease?

A
  • Blood film for sickled cells
  • Sickle solubility test
  • Hb electrophoresis
  • High performance liquid chromatography
1686
Q

Clinical features of sickle cell disease

A

Neuro
- Meningitis, stroke
Retinopathy
- Retinal infarction and glaucoma
Pulmonary HTN
- ACS
- Acute pain event
CV
- Cardiomegaly
- Diastolic heart failure
Haem
- Anaemia
- Leukocytosis
- Asplenia -> splenic infarction, sequestration
Renal
- Papillary necrosis due to albuminuria, isothenuria, substantial kidney injury
Liver
- Gallstones
- Indirect hyperbilirubinaemia
Other
- Priapism
Avascular necrosis

1687
Q

Complications of sickle cell disease

A

Complications
ACUTE
- Acute pain, anaemia, chest syndrome
- Infection
- Stroke
- Priapism
- Aplastic crisis
- Osteomyelitis
- Splenic sequestration
- Dactylitis in children
CHRONIC
- Pain, anaemia
- Sickle nephropathy, retinopathy, hepatopathy
- Leg ulcers
- Pregnancy
- Cardiac - PHT
- Sickle lung disease

1688
Q

Precipitating factors of sickle cell pain/crisis

A
  • Infection
  • Dehydration
  • Stress
  • Hypoxia
  • Temperature
  • Physical exertion
  • Alcohol
  • Unknown
1689
Q

What can help predict if a person with sickle cell disease is going to have sickle pain?

A

May become jaundiced eg eyes may become yellow

May be more irritable or tired than usual

Numbness or sensation change where pain will be

1690
Q

Management of acute chest syndrome in SCD

A
  • Haem emergency
  • O2 sats <94% or PO2<9 on AIR
  • Abx: IV ceftriaxone and PO Clari
  • High flow oxygen/CPAP
  • IVF
  • Incentive spirometry
  • Thromboprophylaxis
  • Regular monitoring /discussion with ITU
  • Exchange transfusion
1691
Q

What is acute chest syndrome in SCD?

A

Acute chest syndrome
1. S/S of respiratory disease (chest pain, fever, wheeze, SOB, cough)
2. New radiological evidence of consolidation

1692
Q

What is the link between Malaria and infection in SCD?

A
  • Malaria: increased morbidity and mortality -> give anti-malarial prophylaxis and ITNs in all endemic areas, prompt diagnosis and treatment
  • Bacterial: encapsulated organisms eg Strep pneumoniae: need Pneumococcal vaccine every 5 years, Hib vaccine, Meningococcal vaccine (A and C), Pen V prophylaxis either until 18 or lifelong
  • Parvovirus: aplastic anaemia
1693
Q

What is the link between sickle cell disease and stroke? How do you manage and prevent it?

A

SCD children at >200x risk of stroke compared to peers
11% of HbSS have stroke by age 20

Management
- Haem emergency
- Exchange blood transfusion
- Identify precipitating cause
- MDT + ICU
Prevent
Investigate high risk or silent infarcts eg
- Transcranial doppler USS
- Neuroimaging
- Neurocognitive assessment
- Psychosocial assessment
Prophylactic therapy with hydroxyurea, chronic exchange blood transfusion

1694
Q

Cause and epi of African trypanosomiasis

A

36 countries in SSA
~55 million people at risk
- T.brucei gambiense: West Central Africa
- T.brucei rhodesiense: East Africa

Uganda has overlap between the two
Cases coming down due to sustained efforts and screening
Disease of rural reas eg Angola, DRC, South Sudan (used to be 30-50% prevalence)

1695
Q

WHO targets for African trypanosomiasis

A

WHO targeted elimination of HAT “as a public health problem” by 2020
…an annual incidence <1 per 10,000 individuals in 90% of endemic areas
…and a global incidence of <2000 per year

New target…
Elimination of gambiense HAT transmission – zero reported cases by 2030

1696
Q

Life cycle for African Trypanosomiasis

A
  1. Infected tsetse fly takes blood meal from mammalian host
  2. Injects metacyclic trypomastigotes into skin -> lymphatics -> bloodstream
  3. Develop into trypomastigotes and replicate via binary fission, parasites move to other sites
  4. Tsetse fly takes meal and ingests trypomastigotes
  5. In the fly’s midgut, the parasites transform into procyclic trypomastigotes, multiply by binary fission
  6. Leave the midgut, and transform into epimastigotes
  7. Epimastigotes reach the fly’s salivary glands continue multiplication by binary fission and transform into met cyclic trypoamastigotes
  8. Repeats

The cycle in the fly takes approximately 3 weeks.
Rarely, T. b. gambiense may be acquired congenitally if the mother is infected during pregnancy.

1697
Q

How can you distinguish T.b.rhodesiense from T.b.gambiense in early stages of infection?

A

50% of people with T.b.rhodesiense will have chancre at bite site

1698
Q

Clinical presentation of African trypanosomiasis

A

Two phases
1) Early stage = haemolymphatic stage
- T.b.rhodesiense: chancre at bite site in 50% of cases, heals often leaving altered pigmentation
- Gambiense: enlarged LNs, spleen, local oedema, cardiac abnormalities, general malaise, headaches, undulating fever
○ Winterbottom’s sign = enlarged neck gland

2) Late stage = encephalitic
- Parasites invade internal organs including CNS
- Gambiense: months/years
- Rhodesiense: 1-4 weeks
Headaches, altered sleep, personality changes, mental function impaired, weight loss, coma and death

1699
Q

Diagnosis of African trypanosomiasis

A

Direct demonstration of parasite
- Blood films eg thick or thin
- Chancre or LN if acute infection
- Blood concentration techniques eg DEAE anion exchange column, microhaemocrit (buffy coat), quantitative buffy coat

LP and CSF examination
- Raised WCC in CSF suggest stage 2

Serology
- Card agglutination test for trypanosomiasis (CATT)
* Drop of blood and antigen (fixed parasites) on plastic card -> blue granular deposits mean infection
Only works for T.b.gambiense but V cheap

1700
Q

Signs and symptoms of scorpion sting

A

Release of ANS mediators = autonomic storm
* Parasympathetic (cholinergic)
○ Vomiting, sweating, hypersecretion, pancreatitis, priapism
* Sympathetic (adrenergic)
○ Goose bumps/flesh, tachycardia, CV disturbances, myocarditis, pul oedema, hyperglycaemia

Neurotoxic effects
- Fasciculations
- Tonic-clonic spasms “pseudo-convulsions”
- Nystagmus
- Opsoclonus
- Irritability
- Paralysis
Ptosis

1701
Q

Prevention of scorpion stingsv

A

Wear gloves and don’t put fingers in at risk areas,
wear long boots and trousers,
and socks in sand or undergrowth,
shake-out clothes,
shoes and backpacks before putting them on,
check living spaces with UV light,
sleep under perthrin impregnated bed net and off ground

1702
Q

Signs and symptoms of cutaneous (dermonecrotic) loxoscelism

A

Loxosceles spider eg american or brown recluse. First 24hr: pain, burning, itching, swelling etc,
48hr: red-white-and-blue sign, blisters, then systemic s/s eg malaise/fever etc,
by 7 days necrotic eschar develops sloughing to leave necrotic ulcer which may need surgery/Abx

1703
Q

Signs and symptoms of cutaneous-haemolytic (visceral) loxoscelism

A

Fever, haemolytic anaemia, jaundice, resp distress, collapse and AKI

1704
Q

Signs and symptoms of wandering spiders eg Latin American wandering/banana spiders

A

Immediate local pain and s/s, then tachycardia, HTN causing stroke, agitation, vomiting, bradycardia, hypotension, arrhythmias, acute pul oedema.

Bite may be dry.
Death by paralysis or asphyxiation.
Give antivenom and supportive care.

1705
Q

Signs and symptoms of envenoming by tarantulas

A

Pain, swellling, persistent + frequent tetanic muscle cramps/spasms in bitten limb then generalised, mild rhabdo

1706
Q

Treatment of spider bites

A

First Aid: hospital and analgesia (pressure-immobilisation for Sydney funnel-web).
Local intense pain: Phoneutria (Brazil) 2% lidocaine then IV dipyrone, Latrodectus (USA): antivenom, Australia: antivenom.
Hospital: Antivenoms for Phoneutria, Atrax, Lactrodectus. antivenom has been used for loxosceles but effectiveness uncertain.

No evidence for supportive tx but can give eg neurotoxic: Ca gluconate, Necrotic: Abx, steroids, surgery etc

1707
Q

Prevention of spider bites

A

Protective clothing, footwear and gloves,
don’t leave gloves or clothing on ground,
apply repellent eg DEET to exposed areas of skin,
watch out for spiders webs,
watch where you put your hands,
flick spiders off skin (don’t crush)

1708
Q

What is a hymenoptera sting and how do you manage it?

A

Eg ants, bees, wasps, hornets. Can be fatal anaphylaxis eg in brazil due to acquired IgE-mediated-hypersensitivity.
Ix: hx, plasma cell mast tryptase, venom-specific IgE (RAST), molecular allergic diagnostics.
Tx: adrenaline if anaphylaxis.
Prevent: avoidance, self-injectable adrenaline and allergy bracelet, desensitisation.

1709
Q

What are lepidoptera and effects of venom? Mx/prevention?

A

Hairy caterpillars/moths. Eg giant silk worm moth catepillar.

PATHOLOGY
Venom has anticoagulant effects -> haemorrhage, haemolysis, renal failure (silkworm caterpillar). Local pain, inflammation, blistering (other species).

Mx: antihistamines, corticosteroids, analgesia, specific antivenom for silkworm caterpillar.

1710
Q

S/S of Ciguatera poisoning and management

A

Within 6-8 hours: GI upset
Then paraesthesias, hot/cold sensation ie cold allodynia/paradoxical dysaesthesia, weakness, ataxia, myalgia

CV: hypotension and bradycardia, hypovolaemia

Management:
IV mannitol early, correct acid-base and electrolyte abnormalities, neuropathic for ongoing symptoms

1711
Q

Scombroid poisoning
- S/S
- Management

A

Immediate peri-oral tingling/pepper sensation
<1hr after ingestion - anaphylactic type symptoms eg rash, pruritic and urticaria, headache, bronchospasm, dizziness and syncope

Histamine, taurine release by enteric bacterial histidine decarboxylase from decaying fish flesh (can be potentiated by histaminase inhibitors eg isoniazid)

Management:
- Adrenaline if anaphylaxis
- Histamien H1 and H2 receptor blockers

1712
Q

Management of marine poisoning

A

If ingestion in 1-2h -> eliminate gut contents
Repeated doses of activated charcoal
Hypotension -> correct hypovolaemia, atropine for bradycardia

1713
Q

Characteristic of severe shellfish poisoning

A

Paralytic: descending paralysis progressing to fatal resp paralysis

Amnesic: short term or permanent amnesia (eg mussel poisoning)

NB: tetradotoxins (eg puffer fish) also cause rapid generalised paralysis

1714
Q

What are the implementation challenges to reducing TB treatment to 4 month regimen?

A

Susceptibility testing
* Need universal DST (minimum RIF and INH; FQ if high incidence)

Directly observed treatment
* DOT for at least 5 days/wk; may not be possible in programmes

Pill burden
* High (because of rifapentine; no fixed-dose combination)

Cost
* Higher than RHZE

Administration with food
* Need to give with at least modest size meal

Programmatic considerations
* Training of HCW, availability of RPT

1715
Q

Two key species of cryptosporidia that cause the most human infections? Relevant treatment?

A

Cryptosporidium parvum
Cryptosporidium hominis

TREATMENT
Most self-limiting in immunocompetent. No current treatment in immunocompromised has been proven to work - paromomycin, nitazoxanide has been trialled.

1716
Q

How long is infection with toxoplasma?

A

Lifelong

1717
Q

How is toxoplasma spread?

A

Ingestion of Undercooked or Contaminated Meat (contained oocysts)

Contact with Contaminated Cat Feces

Contaminated Water or Food

Congenital Transmission

Organ Transplantation or Blood Transfusion

Rarely, individuals can become infected by accidentally inhaling oocysts, especially in areas with poor hygiene or heavy cat populations.

1718
Q

What three forms can infection with toxoplasma take?

A

1) Infections in post-natal, immunocompetent persons

2) Congenital infections

3) Severe and very dangerous disseminated infections in immunocompromised

1719
Q

Relationship between toxoplasmosis and pregnancy?

A

Across trimesters, risk of transmission increases but risk of symptoms/complications decreases. ie risk of transmission lowest in first trimester but highest risk of miscarriage, stillbirth, congenital malformations. However, in third trimester risk of transmission up to 80% but effects on foetus milder and might not be seen until later in life, eg vision loss or learning difficu

1720
Q

Diagnostic investigations for toxoplasma?

A

Anti-toxoplasma IgG

Ag detection via PCR

1721
Q
A
1722
Q

Yaws public health strategy

A

Mapping of foci
• Treatment of whole
community
• Likely need several rounds
• Then mop up with treatment cases & contacts
• Once no more cases
switch to serological surveillance

1723
Q

Yaws public health strategy

A

Mapping of foci
• Treatment of whole
community
• Likely need several rounds
• Then mop up with treatment cases & contacts
• Once no more cases
switch to serological surveillance