Anti-Neoplastic Drugs and Cancer Treatment Flashcards

1
Q

Goals of cancer treatment

A
  1. Cure when possible
  2. Useful prolongation of life where cure is not possible
  3. Relief of symptoms whether cure or life prolongation is the goal
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Methods of cancer treatment

A
  1. Surgery
    - diagnostic –> biopsy to define the existence of tumor
    - curative –> remove all the tumor
    - palliative –> relieve symptoms
  2. Radiation therapy
  3. Chemotherapy
    - cytotoxic
    - targeted
  4. Immunotherapy
    - cytokines
    - antibodies
    - immune cells/vaccines
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What is radiation therapy?

Aim of radiotherapy?

A

What is radiation therapy?
- the treatment of disease, primarily malignant tumors, using ionizing radiation = high energy x-rays or particle radiation

Aim of radiotherapy?
- to deliver as uniform a dose as possible to an accurately localized target, with a goal to kill the tumor cells but avoiding as much normal tissue as possible in order to minimize physical, physiological and psychological consequences for the patient

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

What is radiation?

A

Propagation of energy over some distance from a central location

  • difference between ionizing and non-ionizing radiation is the amount of energy delivered by the radiation
  • primary property of ionizing radiation is to break bonds –> most important target is DNA
  • ionizing radiation = x rays, gamma rays
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

How does radiotherapy work?

A

When ionizing radiation interact with living matter, energy is transferred from the beam

  • biological effects may be direct or indirect (via the formation of free radicals)
  • effects lead to…
  • –> cell death (requires high doses)
  • –> cell mitosis being delayed or cell may die in attempt
  • –> permanent modifications to chromosomes - passed onto daughter cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Radiobiology

A

Critical organelle in the cell is DNA –> when cells are irradiated the following may happen:

  • no effect
  • sublethal damage –> DNA may have breaks but can be repaired
  • lethal damage –> critical targets in cell affected, leads to cell death

Cells are most vulnerable to damage during the mitosis phase of the cell cycle

  • radiotherapy takes advantage of the fact that cancer cells do not behave normally –> due to rapid proliferation, more cells are undergoing proliferation at any given time than the normal cell population
  • cancer cells are most likely to be damaged by radiation and die
  • normal cells are more likely to be able to repair sub-lethal damage and repopulate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Radiation absorbed dose

A

Gray –> amount of energy imparted to matter from any type of radiation

  • Gy = J/Kg
  • Gy = 100 cGy
  • 1 cGy = 100 ergs per gram = 1 rad
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Radiobiology of cancer

A

DNA double strand breaks are most important lesions caused by radiation

  • 2 DSBs may result in cell kill at the time of cell division, mutation, or carcinogenesis
  • doses of radiation are ultimately tied to the elicitation of cell death

DNA damage is the result of direct and indirect effects of radiation

  • DSB is the most important lesion
  • indirect action = photon hits an electron –> results in formation of a free radical that then causes a break in the DNA
  • direct action = electron itself acts as an ion –> directly damages DNA

Damage/Gy of X rays

  • 40 DSB breaks –> most important
  • 150 DNA crosslinks
  • 1,000 SSB –> poorly correlates with lethality
  • 2,500 base damages
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Benefits of dose fractionation

A
  1. Allows repair of sublethal damage –> spares late responding normal tissue preferentially
  2. Reassortment/redistribution of cells in the cell cycle –> increases tumor damage, no effect on late responding normal tissue
  3. Reoxygenation –> increases tumor damage by production of free radicals; no effect in normal tissue
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Normal tissue tolerance

A

There are intrinsic differences in normal tissues in respect to their tolerance of radiation

  1. High sensitivity –> cell turnover is high
    - thyroid
    - lungs
    - breasts
    - stomach
    - colon
    - bone marrow
  2. Intermediate sensitivity
    - brain
    - esophagus
    - liver
    - small intestine
    - ovaries
    - pancreas
    - lymph nodes
  3. Low sensitivity
    - skin
    - dense bone
    - spleen
    - gall bladder
    - kidneys
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Factors that impact chances of long term toxicity from radiation therapy

A
  • high dose
  • high fraction size
  • larger volume treated
  • nature of cells treated
  • overall organization of the tissue
  • –> higher oxygen content (lung)
  • –> proliferative component (GI tract, marrow)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Side effects of radiation

A

Acute and late sequelae –> if someone received radiation in a certain area in the past, you have to consider all of the potential side effects that could occur in the future due to the radiation

Effects on normal tissue are complex –> ie skin:

  • acute erythema
  • erythema, epilation, desquamation can occur after 2-3 weeks
  • re-epitheliazation takes ~6-8 weeks
  • late effects = atrophy, fibrosis, necrosis, stenosis, telangiectasia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

X ray and electron beam dose profiles

A

Different energy radiation give different doses to different tissue depths

  • X rays = have different degrees of penetration at different energies
  • protons = tend to have a very different deposition pattern of dose
  • –> will penetrate between 10-15 cm before delivering most of their dose
  • –> most of their dose and the width of the deposition is much smaller than X rays = underlies the use of protons where you want to minimize the impact on adjacent structures

Gradually decrease the field of radiation over subsequent treatment

  • want to make sure that the initial field is large enough to target outreaching portions of the tumor that may not be obvious by radiology
  • final dose is an intense, targeted “boost” to get the last remaining difficult portions of the tumor
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Radiation delivery methods

A
  1. External beam radiotherapy –> tele-therapy
  2. Brachytherapy –> insert radioactive source close to tumor
  3. Systemic radiotherapy –> administer isotope to circulation (e.g. isotopes of iodine for thyroid cancer)
  4. Radio-immunotherapy –> attach to antibody
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Overview of cancer chemotherapy

A

Chemotherapy = use of drugs to treat cancers

  1. Cytotoxics –> more toxicity to tumor cells than host within a dose range
    - chemicals
    - natural products –> extracts of plants, bacteria, animals
  2. Targeted agents –> modulate tumor cell biology to decrease viability
    - hormonal/receptor agonists/antagonists
    - oncogene products
    - growth factor receptors
    - immune modulators –> cytokines, antibodies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

General principles of cytotoxics

  • Log cell kill
  • Combination chemotherapy
A

Log cell kill:

  • each dose of drug kills a constant fraction of cells –> results in decrease by a number of “logs” of cell numbers
  • successive cycles of chemotherapy necessary to get down to a low number of ideally poorly growing tumor cells
  • time each round of therapy with recovery of host from toxicity
  • better effect with smaller initial tumor volume + as high of a dose as possible

Combination chemotherapy:

  • use of more than one drug –> each drug of regimen ideally have non-overlapping host toxicities
  • each drug in a combination regimen has activity on its own
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

2 mechanisms of cell death in chemotherapy

A
  1. Induction of DNA damage that causes mitochondrial activation of apoptosis via caspase 3
  2. Elaboration of a cell death program via a death signal acting on a receptor
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Alkylating agents

A

Act to cause covalent modification of cellular molecules with alkyl moieties

  • different classes of agents differ in spectrum of cellular molecules affected –> all modify DNA structure
  • intra/inter-strand crosslinks have greatest value as ant-cancer agents
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Major toxicities of alkylating agents

A
  • myelosuppression –> in general there is a return to normal levels of leukocytes and platelets between 18-22 days allowing for another round of treatment
  • GI epithelial damage = mucositis
  • alopecia
  • impaired wound healing
  • impaired growth in children
  • reduced resistance to infection
  • nausea and vomiting
  • sterility
  • teritogenic
  • carcinogenic
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Busulfan

A

Alkylating agent - an alkyl sulfonate

  • causes acute and delayed myelosuppression
  • lung damage with high doses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Cyclophosphamide

A

Alkylating agent

  • bladder toxic in high or prolonged oral dosing
  • high doses –> cardiotoxicity, SIADH, lung toxicity
  • requires hepatic activation –> breakdown products are toxic to the bladder
  • Use MESNA and hydration to protect bladder with high doses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Ifosfamide

A

Alkylating agent

  • requires hepatic activation –> metabolites also bladder toxic and cause encephalopathy
  • routine use of hydration and MESNA required
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Procarbazine

A

Alkylating agent

  • oral bioavailability
  • CNS penetration –> CNS toxic effects = somnolence, mood swings
  • part of curative regimens in hodgkin’s lymphoma
  • current palliative use in gliomas (brain tumors)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

DTIC/Temozolamide

A

Alkylating agents –> both activated to the same intermediate

  • alkylate O6 of guanine
  • distinct repair pathway
  • alkylguanine alkyl transferase
  • DTIC –> active in melanoma
  • Temozolomide –> oral active in glioma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Nitrosureas

  • Carmustine (BCNU)
  • Lomustine (CCNU)
A

Bifunctional alkylating agents

  • wide spectrum of use
  • cross blood/brain barrier
  • active agent against CNS metastases and brain tumors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

Platinum derivatives

  • Cisplatin
  • Carboplatin
  • Oxaliplatin
A

DNA damaging heavy metals –> act as alkylating agents

  • bind N-7 of guanine on single strand
  • cross links DNA
  • binds proteins
  • Cisplatin –> requires hydration = IMPORTANT
  • Carboplatin –> dosed according to the patient’s renal function

Toxicities differ:

  • cisplatin = renal > neuro/oto > heme
  • carboplatin = heme&raquo_space;» renal/neuron
  • oxaliplatin = neuro&raquo_space; heme&raquo_space;» renal
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Anti tumor antibiotics

A

Historically found to be produced by bacteria and have anti-bacterial and anti-tumor cell activities

  • bind to DNA and cause damage to physical structure of DNA or alter DNA function
  • important drugs = bleomycin + actinomycin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Bleomycin

A
  • oxygen increases toxicity of bleomycin
  • renal clearance
  • tissues clear by a bleomycin hydrolase –> lungs + skin lack the hydrolase –> causes pulmonary and cutaneous toxicity, especially with decreased renal function
  • increased toxicity with cumulative dose
  • toxicity worse with underlying pulmonary disease
  • persists in lung –> fatal activation of lung toxicity during surgery with high oxygen inspired
  • cause Raynaud’s hypersensitivity
  • part of curative regimens for germ cell and hodgkins disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Actinomycin D

A

Intercalate into DNA helix –> inhibit RNA/protein synthesis
- part of curative regiment for pediatric cancers

Toxicities

  • myelosuppression
  • nausea/vomitting
  • mucositis
  • diarrhea
  • vesicant –> avoid extravasation
  • radiation recall
30
Q

Topoisomerase directed agents

A

Topoisomerases allow unwinding of coiled DNA molecules

  • bind to DNA to make a break
  • topo I = makes a single break –> forms an enzyme-DNA intermediate –> reseals the link after one strand rotates around the other
  • topo II = makes a double strand break –> enzyme binds to broken ends –> passes another whole double helix between the strands

Topo-directed drugs form a ternary complex = drug + enzyme + DNA –> stabilizes the break + signals DNA repair and damage responses

Specific agents:

  • Topo I = topotecan, irinotecan (derivatives of camptothecin)
  • Topo II = doxorubicin, daunorubicin, idarubicin (anthracyclines), mitoxantrone, etoposide
31
Q

Anthracyclines

A

Doxorubicin, daunorubicin, epirubicin, idarubicin

Toxicities:

  • myelosuppression, mucositis, alopecia, cardiac toxicity –> dose related
  • radiation sensitive
  • extravasation damage to surrounding tissues - vesicant
  • idarubicin = somewhat less cardiotoxic on a weight basis
  • eliminated prominently by liver –> dose adjustment needed in hepatic failure
  • generate free radicals –> not related to topo II –> causes cardiotoxicity
32
Q

Mitoxantrone

Etoposide

A

Mitoxantrone –> similar structure to anthracyclines but less cardiotoxic and emetogenic

Etoposide (aka VP-16)

  • toxicities = neutropenia, thrombocytopenia, alopecia
  • reduce dose in proportion to creatinine clearance
33
Q

Topotecan

Irinotecan

A

Topo I inhibitors

Topotecan
- toxicites = nausea, myelosuppression, fatigue, alopecia

Irinotecan

  • toxcitiies
  • –> early onset diarrhea = atropine sensitive
  • –> late onset diarrhea = reflects enterohepatic clearance
  • –> myelosuppression
  • –> alopecia/nausea/fatigue
34
Q

Anti-metabolites

  • Folate antagonists
  • Pyrimidine analogs
  • Purine analogs
A

Folate antagonists

  • methotrexate
  • pemetrexed

Pyrimidine analogs

  • fluorouracil
  • cytarabine
  • gemcitabine
  • azacytidine/deazacytidine

Purine analogs

  • thioguanine
  • mercaptopurine
  • pentostatin
  • fludarabine
  • cladribine
35
Q

Common features of antimetabolite action

A
  • cells must be in S phase to be maximally active –> must be growing cells
  • mimic normal nucleic acid precursors and can be misincorporated into DNA or RNA –> results in altered function of DNA or RNA –> causes cell death
  • interfere with production of DNA and RNA precursors –> inhibits normal nucleotide synthesis
  • longer the exposure, the most toxic to the host –> more cells enter S phase and become effected
  • may be able to rescue by replacing downstream metabolite
36
Q

Antimetabolites

  • Inhibitors of ribonucleotide synthesis from purines
  • Inhibitors of deoxyribonucleotide synthesis from ribonucleotides
  • Inhibitors of DNA synthesis from deoxyribonucleotides
A

Inhibitors of ribonucleotide synthesis from purines

  • mercaptopurine
  • thioguanine
  • methotrexate (high dose)

Inhibitors of deoxyribonucleotide synthesis from ribonucleotides
- hydroxyurea

Inhibitors of DNA synthesis from deoxyribonucleotides

  • methotrexate (low/high dose)
  • cytarabine
  • 5-flurouracil
  • asparaginase (indirect)
37
Q

Methotrexate

  • mechanism
  • major toxicity of standard dose MTX
A

Mechanism = inhibits dihydrofolate reductase, blocking DNA and RNA precursor formation

Major toxicity of standard dose MTX = myelosuppression

  • other prominent = mucositis
  • chronic exposure = hepatic fibrosis
38
Q

High dose MTX

A

High dose MTX = give intentionally lethal dose

  • use leucovorin = downstream metabolite –> already reduced folate does NOT require dihydrofolate reductase for action after exposure to high dose MTX
  • leucovorin rescues normal cells while “loading” cancer cells with high concentrations of MTX
  • cancer and normal cells have different folate receptors which accounts for differential uptake of leucovorin
39
Q

5-fluorouracil

  • mechanism
  • major toxicities
  • metabolism
A

Mechanism = inhibits thymidine synthesis by forming covalent complex with folate and thymidylate synthase
- orally available pro-drug = capecitabine

Major toxicities

  • myelosuppression –> more continuous with infusion
  • GI –> especially bolus 5FU
  • cerebellar/neurocognitive
  • coronary spasm –> rare

Metabolism

  • metabolized in tissues by DHPD –> useful when there is hepatic/renal impairment
  • deficiency of DHPD = severe toxicity

Enhance toxicity/activity of 5FU by coadministration with reduced folate –> usually leucovorin

40
Q

Leucovorin uses

A
  1. Rescue/protect normal cells after high doses of MTX
    - can treat MTX toxicity encountered even at lower doses
  2. Enhance 5FU toxicity to cancer cells by increasing formation of ternary complex of thymidylate synthase + folate + 5FU
41
Q

Cytosine analongs

  • major toxicity
  • metabolism
A

Cytarabine + gemcitabine

Major toxicity = myelosuppression, esp after continuous venous infusion
- cerebellar, eye irritation, hand foot

Metabolism
- metabolized by cytidine deaminase –> useful in hepatic/impairment

Bolus regimens of cytarabine (AraC)
- rationale = drive production of AraCTP

Continuous venous infusion regimens = prolong exposure –> maximize S phase exposure

Gemcitabine –> used in solid tumors, unique toxicity = hand food prominent

42
Q

Purine analogs: Bases

  • Mechanism
  • Metabolism
  • Toxcities
A

6-Mercaptopurine + 6-Thioguanine

Mechanism

  • inhibition of de novo purine synthesis
  • some incorporation into DNA (6MP) = dysfunction of DNA
  • incorporation into RNA
  • must be anabolized to nucleotide for incorporation

Metabolism of 6-mercaptopurine

  • metabolized by xanthine oxidase –> use with allopurinol (drug to control uric acid synthesis) requires dose reduction
  • thiopurine methyltransferase –> genetic deficiency results in excessive toxicity

Toxicities = myelosuppression, nausea, rare hepatotoxicity

43
Q

Purine analogs: nucleosides

  • Mechanisms
  • Toxicities
A

Fludarabine = prototypic of adenine based analogs
- also cladribine

Mechanism

  • incorporate into DNA as false nucleotide
  • inhibit DNA synthetic activities

Toxcities

  • myelosuppresion
  • immunosuppression –> results in infections
  • neurotoxicity at high doses
  • rare interstitial pneumonitis and rare hemolytic anemia

Pentostatin unique –> not directly incorporated into DNA but inhibits adenosine deaminase –> therefore builds up levels of dATPs = signal to apoptosis in T cells

44
Q

Asparaginase

  • mechanism
  • toxicities
A

Mechanism

  • asparaginase = foreign protein that clears circulation of asparagine
  • consequence –> decreased protein synthesis in susceptible cell types
  • DNA synthesis requires concomitant protein synthesis –> therefore indirectly damages DNA

Toxicities

  • hypersensitivity
  • hyperglycemia
  • pancreatitis
  • altered clotting functions
45
Q

Hydroxyurea

A

Mechanism

  • reversible inhibition of ribonucleotide reductase by chelation of non-heme Fe at reactive center
  • consequence –> decreased dNTP pools with consequent stalled replication forks –> perceived by the cells as damaged DNA and a signal for apoptosis

Toxicities

  • myelosuppression
  • mucosal damage
  • enhanced effect in renal failure

Oral bioavailability

46
Q

Microtubule directed agents

  • mitotic inhibitors
  • microtubule polymer stabilizers
  • toxicities
A

Mitotic inhibitors

  • vinca alkaloids = vincristine + vinblastine
  • vincristine = neurotoxic > myelotoxic
  • vinblastine = myelotoxic > neurotoxic

Microtubule polymer stabilizers –> promote microtubule formation, but microtubules formed are abnormal and shortened –> don’t function normally –> prevent cell cycle progression through M phase

  • taxanes = paclitaxel, docitaxel
  • epothilones

Common toxicities

  • myelosuppression
  • neuropaty
  • fluid retention

Taxane specific side effects

  • fluid retention/vascular leak
  • hypersensitivity reactions
47
Q

Arsenicals

A

Arsenic trioxide

  • uniquely toxic to acute promyelocytic leukemia
  • generates free radicals

Toxicities
- heavy metal toxicity = kidney + cardiac conduction (prolongs the QT interval)

48
Q

Thalidomide derivatives

A

Unique cytotoxicity for developing endothelial cells = basis for teratogenic effects

  • interact with cytokine production and elaboration
  • useful in certain hematopoeitic neoplasms by mix of cytotoxic and immunoregulatory mechanisms

Toxicities

  • teratogenic
  • neuropathy
  • cytopenias
  • GI
49
Q

Targeted therapy in cancer treatment

A

Drugs directed at the molecular or physiologic concomitants of malignancy

In contrast to cytotoxics…

  • saturability of dose-response
  • biological effective dose rather than maximal tolerate dose
  • patient selection possible by presence of the drug target

Most targeted therapies target nuclear steroid hormone receptors –> all are transcription factors

50
Q

Steroid hormone receptor agonists/antagonists –> Prostate cancer

A

Androgen receptor modulation

LHRH analogs –> decrease LH release by pituitary = decreased testosterone production

Antiandrogens = act at level of androgen receptor

  • flutamide
  • casodex

High dose estrogen –> feedback inhibition in hypothalamus to decrease LH elaboration

51
Q

Steroid hormone receptor agonists/antagonists –> Breast cancer/endometrial cancer

A

Estrogen/progesterone receptor modulation

Antiestrogens –> tamoxifen

Aromatase (estrogen synthetase) inhibitors

  • letrozole
  • arimidex
  • examestane

Progestational agents

52
Q

Steroid hormone receptor agonists/antagonists –> Leukemia

A

Glucocorticoid receptor action induces apoptosis in lymphoid cells

Glucocorticoids –> high doses of prednisone, prednisolone and dexamethasone

53
Q

Aromatase inhibitors

- mechanism of action

A

Type I = steroidal inhibitor –> target the substrate binding site

Type II = nonsteroidal inhibitor –> target the cyt P450 aromatase

54
Q

All trans retinoic acid

  • mechanism
  • toxicities
A

Mechanism = binds to retinoic acid receptor
- induces differentiation of leukemic cells in acute promyelocytic leukemia that have a 15:17 translocation that alters the structure and function of the retinoic acid receptor

Toxicities

  • teratogenic
  • cutaneous = dry skin, ocular keratitis
  • increased intracranial pressure
  • differentiation syndrome
55
Q

Tyrosine kinases

A

Oncogenes act through protein kinases

TKs = transfer phosphate from ATP to substrate tyrosines

Integral membrane TKs

  • respond to external signals –> EGFR, PDGF, IGF
  • respond to “matrix/contact” –> integrins
  • dimerize to signal –> Her2/neu

Non-integral membrane TKs

  • associate with primary signal recipients –> Ick, lyn, src
  • may act independently –> abl
  • may associate with membrane by post translational modification –> myristoylation
56
Q

Common elements/repeated themes with TKs

A
  • overexpressed or activated in cancer –> ex = EGFR (lung cancer) + Her2/neu (breast cancer)
  • altered activity by mutation –> ex = c-kit
  • altered activity by translocation –> ex = bcr-abl
  • overexpression associated with advanced stage + inferior prognosis
57
Q

Targeted cancer therapy - ATP site protein kinase antagonists
- common toxicity patterns

A

Since all protein kinases have structural similarities to ancestral kinases, ATP antagonists have varying degrees of shared capacity to inhibit many different families of kinases –> more specific the binding to one kinase, the more specific the inhibitor action
- results in common toxicity patterns

Bcr/abl

  • cytopenias
  • liver abnormalities
  • fluid retention
  • rare cardiomyopathy

VEGF receptor antagonists

  • hypertension
  • proteinuria
  • perforated viscus
  • clotting/bleeding

EGFR/ERBB2

  • Diarrhea
  • cutaneous
58
Q

Targeted cancer therapy - MTOR protein kinase antagonists

- common toxicity patterns

A
  • stomatitis
  • thrombocytopenia
  • hyperlipidemia
59
Q

Targeted cancer therapy - post-translational modification of chromatin proteins/DNA methylation

A

Interact with epigenetic regulation of gene expression

  • histone acetylation
  • DNA methylation
  • –> methylated cytosines cause transcriptional repression
  • –> hypomethylation “wakes up” repressed genes and allows differentiation

Low doses of certain nucleosides inhibit cytosine methylation

60
Q

Modulators of chromatin function

A

Histone deacetylase inhibitors

  • vorinostat
  • romidepsin

DNA hypomethylating agents –> low doses of certain nucleosides

  • azacytidine
  • decitabine
61
Q

Proteasome inhibitors

A

Bortezomib
- clinically active in multiple myeloma and lymphomas

Major toxicites

  • neuropathy
  • thrombocytopenia
  • altered GI tract function
62
Q

Biological response modifiers

  • IFNs
  • IL-2
A

IFNs

  • adjuvant for melanoma
  • hairy cell leukemia
  • kaposi’s sarcome
  • side effects: fatigue, ctyopenias, fevers, chills

IL-2

  • induces T cell response –> cytolytic for tumors
  • renal, melanoma
  • high dose IL-2 produces complete responses in small subset of metastatic renal tumors
  • side effects: same as IFNs + hypotension, vascular leak, altered mental status, cardiopulmonary
63
Q

Biological response modifiers

- Immune modulators = “imis”

A

Thalidomide + lenalidomide

  • alter cytokine milieu that promotes angiogenesis + cell growth
  • uniquely active in multiple myeloma and certain pre-leukemia syndromes

Side effects

  • thromboses
  • edema
  • cytopenias
  • fevers/chills
64
Q

Cetuximab + Panitumumab

A

Anti-EGFR

  • colon cancer with chemo in ras allele wild type
  • squamous head and neck with radiation

Toxicities:

  • infusion reaction –> fever, chills, rare hypotension, bronchospasm
  • cutaneous
  • diarrhea
65
Q

Trastuzumab (herceptin)

A

Anti- Her2/Neu

  • inhibits signaling and immune action
  • hormone independent breast cancer

Toxicities

  • infusion reaction
  • cardiomyopathy
66
Q

Rituximab

A

Anti CD20 on B cells

  • non-Hodgkins lymphoma
  • acts by inhibiting lymphocyte signaling and immune activation

Toxicities
- infusion reaction

67
Q

Anti-angiogenic agents

A

Bevacizumab = anti-VEGF monoclonal antibody

  • active in tumors that prominently express VEGF = renal + glioma
  • potentiates actions of chemo in tumor by acting on microvasculature to decrease tumor interstitial pressure

Toxicities

  • hypertension
  • clotting
  • bleeding
  • perforation of viscus
  • CNS dysfunction
68
Q

Immune cell approaches to kill tumor cells

A

Ipilimumab = anti-CTLA4 antibody

  • CTLA4 and PD1 are “negative” signals to T cell activation
  • approved for treatment of melanoma

Side effects:

  • autoimmune like side effects in gut
  • hepatic
69
Q

Resistance to cancer treatment

A
  • excessive degradation of drug
  • altered affinity of drug target
  • changes in metabolic activation
  • altered drug transport
  • gene amplification of drug target
  • DNA repair mechanisms
  • increase thiol content (alkylators)
  • altered apoptosis induction threshold
70
Q

MDR gene family amplification

A

MDR = multidrug resistance protein = p-glycoprotin

  • normal transport protein
  • energy dependent efflux pump for lipophilic drugs
  • overexpressed in drug resistant leukemia
  • overexpressed in normal and cancer stem cells
71
Q

Limitations of current cancer treatment approaches

A
  • severe toxic effects of many agents
  • lack of selectivity of the drugs against tumor ells
  • lack of knowledge of target expression
  • uncertainty of how target presence convey basis for activity
  • total elimination of malignant cells is usually not possible with therapeutic doses
  • host’s immune response is inadequate to deal with the remaining cells