Principles of Treatment of Pediatric Oncology Flashcards

1
Q

What does a low therapeutic index mean?

A

The ratio between efficacious dose and toxic dose is very small, so small errors in dosing can have big consequences

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

Phase I Trial?

A

Toxicity and determine maximum tolerated dose

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

Phase II Trial?

A

Toxicity and response rate

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

Phase III Trial?

A

Efficacy vs. standard

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

Phase IV Trial?

A

Efficacy, safety, long-term effects after approval

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

Most common type of Phase I trial? Goal? How to determine if dose level is tolerable? Downside?

A
  • 3+3 design: enrol 3 patients in clinical trial, give drug, and assess for toxicity
  • Goal: less than 33% of patients have “unacceptable toxicity” - previously defined, known as “dose limiting toxicity”
  • If no patients have dose limiting toxicity, then go up to next dose level
  • If one patient has toxicity, enrol another 3 patients. If one more patient does have toxicity, then dose level is considered intolerable, and dose is de-escalated. If only the 1/6 or none, go up to next level and repeat same.
  • Downside is have to pause trial while await toxicity assessment, so can become quite lengthy for patients who are waiting
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Rolling 6 design of phase I trial? How to escalate dose?

A
  • Allows you to enrol 6 patients at a time
  • Can escalate as long as you know that 3/3, 4/4, 5/5, 6/6 patients do not actually have toxicity
  • But if more than one has toxicity, have to slow down or de-escalate
  • Useful for multicentre trial with patients waiting
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What is the final dose level called?

A
  • Maximum tolerated dose: <33.3%
  • Is the recommended Phase II dose
  • Europe and US definitions different
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What is a simon 2 stage design for phase II trial?

A

-usually pick a response rate of about 30%
If 1 patient or <1 in first 10 who does not have a response, then will never reach 30% and will close the trial
-But if >3 patients respond then know will reach endpoint and don’t have to go on to second stage
-If have 2-3 who respond, want to expand

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

What is the utility of the principle of multi-drug therapy?

A

To overcome drug resistance to individual agents

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

What is the Goldie-Coldman hypothesis?

A
  • Mathematical model
  • Hypothesis is that cancer cells within a tumour mutate to a resistant phenotype at a rate that is intrinsic to the genetic instability of the particular tumour
  • Will predict that if you use different chemo agents with different mechanisms of action, different toxicities or no overlapping toxicities in setting of MRD, then would increase likelihood of cure in those particular patients
  • This may be less relevant with biologic therapy, but for chemo, forms basis of therapy
  • Other tenant of hypothesis is that don’t give same drugs over again - “trick” cancer by alternating strategies - e.g. alternating cycles
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Principle of dose intensity?

A
  • Theory that maximize dose to kill the most cells, but don’t want to give at cost to patient
  • Why supportive care is important and has contributed to improved outcomes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

For drug disposition in the body - what is ADME? What things affect it?

A
  • Absorption
  • Distribution
  • Metabolism
  • Excretion
  • Clearance, half-life, AUC, volume of distribution, bioavailability and biotransformation
  • Age, organ function, drug interactions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Examples of common pro-drugs?

A

Cyclophosphamide
Ifosphamide
Irinotecan

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

Significance of pharmacodynamics?

A

-Effects of drug on the body, relationship between drug concentration and effect

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

What is pharmacogenomics referring to?

A

Genetic variations effect on spectrum of drug action

e.g. UGT1A1 deficiency

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

Phases of cell cycle?

A

G0 - resting phase, cell has left the cell cycle
G1 - cells increase in size, ready for DNA synthesis
S - DNA replication
G2 - cells increase in size, ready for mitosis
M - mitosis

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

What cell cycle phase do alkylating agents work at?

A

G0

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

What cell cycle phase do antitumor antibiotics work at?

A

Cell cycle non-specific

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

What cell cycle phase do antimetabolites work at?

A

S

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What cell cycle phase do plant alkaloids work at?

A

M

22
Q

What cell cycle phase do hormones work at?

A

Cell cycle non-specific

23
Q

Determinants of CNS penetration?

A

Drug properties

  • Lipophilicity (more lipid avid, better it gets in)
  • Molecular size
  • Degree of ionization
  • Free plasma concentration

Cerebral blood flow

24
Q

Strategies to enhance CNS penetration?

A
  • High-dose systemic chemotherapy - MTX, cytarabine
  • Drugs that penetrate the BBB - nitrosoureas, thiotepa, camptothecins
  • Disrupting the BBB - mannitol (if take cisplatin alone, doesn’t penetrate BBB… one theory is mannitol helps this, another is that BBB not intact in BT patients)
  • Administration of drug into the intrathecal space - MTX, Cytarabine
25
Q

What is the idea of age based intra-thecal dosing based on?

A

After 3 y o volume of CSF is relatively stable

26
Q

Age based intra-thecal dosing of MTX and cytarabine?

A

-<1: MTX 6mg, AraC 15mg
-1: MTX 8mg, AraC 30mg
-2: MTX 10mg, AraC 50mg
>/=3: MTX 12mg, AraC 70mg

27
Q

What does 1 gray mean?

A

The amount of radiation depositing 1 joule of energy in 1 kilogram

28
Q

What factors affect radiation sensitiviy?

A
  • Cell type (mucous membranes, gonadal tissue v. sensitive, bone not as sensitive)
  • Growth and replicative activity
  • Cell cycle phase at the time of exposure (dormant cells more resistant to radiation than those that are actively mutating)
  • Genetic mutations in oncogenes and tumor suppressor genes
  • Microenvironment
29
Q

Examples of disorders with radiation hypersensitivity?

A
  • Ataxia telangiectasia
  • Nijmegen breakage syndrome
  • Fanconi anemia
30
Q

Benefit of proton therapy?

A

Less scatter, prevent damage to sensitive tissues around the tumour

31
Q

What type of radiation therapy is standard for most patients in pediatrics?

A

Intensity Modulated Radiation Therapy (IMRT)

32
Q

What does intensity modulated radiation therapy (IMRT) mean? What is this important for?

A
  • Manipulating radiation to create custom 3D dose distribution
  • Can alter dose distribution to tumour and normal tissue
  • Photons or protons
  • Important for if tumour needs high dose of radiation (e.g. flank rads for WT, don’t need IMRT, or if whole lung rads don’t need IMRT, would just give conformed rad therapy in those areas)
33
Q

Cons of IMRT?

A
  • Complexity increases errors

- Need good Radiation planning to avoid errors in dose calculation, distribution, delivery and machine failure

34
Q

Benefits of Proton Beam Radiation Therapy?

A
  • Favourable dose distribution
  • Spare normal tissue
  • No exit dose leading to sparing underlying normal tissue
  • Potential decrease in entrance dose
  • Total dose is delivered to level of Bragg peak
35
Q

What is brachytherapy? When is it useful? Time frame? Limited by?
Tumour types?

A
  • Uses radioactive implants or molds to deliver local radiation
  • When need to give interstitial, intracavitary or surface rads
  • Time frame 3-5 days
  • Limited by size of tumour bed (if huge tumour bed better off doing external beam)
  • Tumour types: rhabdomycosarcoma (mostly GU, where don’t want to radiate entire pelvis) and used to be used in retinoblastoma
36
Q

What is the Gross Tumour Volume (GTV)?

Difference for photons, protons and brachytherapy?

A
  • The volume occupied by tumour at diagnosis

- Same for photons, protons and brachytherapy

37
Q

What is the Clinical Target Volume (CTV)?

Difference for photons, protons and brachytherapy?

A
  • Includes GTV and sites of suspected occult disease and involved adjacent lymph nodes - include bigger margin due to concern about microscopically positive margins. Have decreased local relapses with this.
  • Same for photons, protons and brachytherapy
38
Q

What is the Planning Target Volume (PTV)? Difference for photons, protons and brachytherapy?

A
  • Includes CTV including surrounding geometric area accounting for variability ini set, breathing and motion during treatment
  • PTV is not the same as photons for protons
  • PTV is same as CTV for brachytherapy
39
Q

What is the CNS Threshold Radiation Dose?

A

Dose at which you begin to see deficits

40
Q

What is the CNS Threshold Radiation Dose?

A

Dose at which you begin to see deficits (table in talk)

41
Q

Special considerations for Peds Rad Onc?

A
  • Growing and developing tissue more sensitive to late effects e.g. RMS of H&N when young
  • Combined with pharmacologic agents (chemoradiotherapy - e.g. radiation recall injury, typically seen with doxo and dactinomycin, so don’t give them together with rads)
  • Technological advances (IMRT, Image guided radiation therapy, stereotactic body radiotherapy, stereotactic radiosurgery)
42
Q

What does the field cover when you give cranial rads for ALL?

A

-Posterior half of eye (optic nerve area) and C2

43
Q

Why do they extend the radiation field to include the whole vertebral body in flank irradiation e.g. for Wilms Tumor?

A

To avoid scoliosis (history of patients with severe after flank irradiation)

44
Q

What is included in whole abdominal radiation if spillage or peritoneal deposits? Why?

A
  • Pelvis

- If limit to just abdomen, can have recurrence in pelvis

45
Q

Because want apices of lungs, what might the field include in classic whole lung radiation?

A

Thyroid gland

46
Q

What is included in spinal radiation? What is it used for?

A
  • Thecal sac, extends to S3
  • Used for metastatic medulloblastoma and other CNS
  • Almost never for CNS leukemia unless refractory
47
Q

What is the most common emergency in patients with hematologic malignancy at diagnosis?

A

Tumor lysis syndrome

48
Q

What diseases is TLS commonly seen in?

A
  • Non-Hodgkin Lymphoma (Burkitt)

- Acute leukemia

49
Q

Lab definition of TLS? Clinical TLS?

A
  • Hyperuricemia: Serum uric acid >ULN in children (>8mg/dL in adults)
  • Hyperkalemia: Serum potassium > 6meq/L
  • Hyperphosphatemia: Serum phosphorus > 6.5mg/dL in children (>4.5mg/dL in adults)
  • Hypocalcemia

Clinical: Renal insufficiency, cardiac arrhythmias, seizures, death

50
Q

Formula for corrected calcium in mg/dL?

A

Corrected calcium in mg/dL = measured calcium in mg/dL + 0.8(4-albumin in gms/dL)

51
Q

Pathophysiology of TLS?

A
  • Cell break down leads to:
  • -Degradation of DNA to purines and phosphates
  • -Release of intracellular potassium
  • -Metabolic conversion of purine analogues to hypoxanthine, xanthine and uric acid by xanthine oxidase
  • -Hyperphosphatemia –> secondary hypocalcemia
  • -Cytokine release causing hypotension and inflammation resulting in renal injury
  • -Increased uric acid and calcium phosphate crystals leading to nephropathy