Cancer Pharmacology Flashcards

1
Q

Categories of alkylating agents used in chemotherapy

A

Bis (chloroethyl) amines, nitrosoureas, aziridines, alkylsulfonate, non-classic alkylating agents, platinum analogs

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

Of the alkylating agents, name the 4 bis (chloroethyl) amines

A

Cyclophosphamide
Mechlorethamine
Melphalan
Chlorambucil

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

Of the alkylating agents, name the 2 nitrosureas

A

Carmustine

Streptozocin

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

Of the alkylating agents, what is the major aziridine used in chemotherapy?

A

Thiotepa

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

Of the alkylating agents, what is the major alkylsulfonate used in chemotherapy?

A

Busulfan

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

3 non-classic alkylating agents

A

Procarbazine
Dacarbazine
Bendamustine

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

Of the alkylating agents, name the 3 platinum analogs

A

Cisplatin
Carboplatin
Oxaliplatin

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

4 categories of antimetabolites used in chemotherapy

A

Antifolates
Fluoropyrimidines
Deoxycytidine analogs
Purine antagonists

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

Of the antimetabolites, what are the 3 antifolates used in chemotherapy?

A

Methotrexate
Premetrexed
Pralatrexate

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

Of the antimetabolites, what are the 3 fluoropyrimidines used in chemotherapy?

A

5-FU
Capecitabine
TAS-102

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

Of the antimetabolites, what are the 2 deoxycytidine analogs used in chemotherapy?

A

Cytarabine

Gemcitabine

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

Of the antimetabolites, what are the 4 purine antagonists used in chemotherapy?

A

6-thiopurines:
6-mercaptopurine (6-MP)
6-thioguanine (6-TG)

Other:
Fludarabine
Cladribine

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

Categories of chemotherapy drugs considered natural products

A

Vinca alkaloids

Taxanes and other anti-microtubule drugs

Epipodophyllotoxins

Camptothecins

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

Of the natural products used in chemotherapy, name the 3 vinca alkaloids

A

Vinblastine
Vincristine
Vinorelbine

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

Of the natural products used in chemotherapy, name the 5 taxanes and other antimicrotubule drugs

A
Paclitaxel
Docetaxel
Cabazitaxel
Ixabepilone
Eribulin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Of the natural products used in chemotherapy, name the epipodophyllotoxin

A

Etoposide

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

Of the natural products used in chemotherapy, name the camptothecins

A

Topotecan

Irinotecan

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

Name the anti-tumor antibiotics

A

Anthracyclines: doxorubicin, daunorubicin, idarubicin, epirubicin, mitoxantrone, derazoxane

Mitomycin (myotmycin C)

Bleomycin

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

5 tyrosine kinase inhibitors used in chemotherapy

A
Imatinib
Dasatinib
Nilotinib
Bosutinib
Ponatinib
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Growth factor receptor inhibitors used in chemotherapy

A
Cetuximab
Panitumumab
Necitumumab
Erlotinib
Afatinib
Osimertinib
Bevacizumab
Ziv-aflibercept
Ramucirumab
Sorafenib
Sunitinib
Pazopanib
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Most widely used alkylating agent with high oral bioavailability, administered by IV or orally, and activated to the 4-hydroxy intermediate

A

Cyclophosphagmide

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

Cyclophosphamide is activated to become 4-hydroxycyclophosphamide, which travels alongside aldophosphamide in circulation. What is the fate of these molecules in liver and tumor tissue?

A

4-hydroxycyclophosphamide is inactivated in liver or tumor tissue (inactive metabolites include 4-ketocyclophosphamide and carboxyphosphamide)

Aldophosphamide cleaves spontaneously in tumor tissue generating the toxic metabolites (toxic metabolites include phosphoramide mustard and acrolein)

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

AEs of cyclophosphamide

A

Phosphoramide mustard responsible for antitumor effects

Acrolein causes hemorrhagic cystitis

Patients should receive vigorous IV hydration during high-dose treatment

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

Cyclophosphamide has broad clinical applications. What are some of them?

A

Essential component of many effective drug combinations for: NHL, other lymphoid malignancies, breast and ovarian cancers, solid tumors in children

Burkitt lymphoma — complete remissions and presumed cures when given as a single agent

Used in combo with doxorubicin an da taxane as adjuvant therapy after surgery for breast cancer

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

First clinically used nitrogen mustard

Most reactive of drugs in its class

Clinical application include topical use for tx of cutaneous T cell lymphoma (CTCL)

Largely replaced by more stable chemotherapeutics

A

Mechlorethamine

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

Melphalan is not a vesicant and has inconsistent absorption orally so it is given by IV. What are adverse effects of melphalan?

A

Toxicity is mostly hematologic

NOTE: N/V and alopecia are less frequent vs. other chemotherapeutics

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

Clinical application of melphalan

A

Primarily used to treat multiple myeloma with dexamethasone

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

Oral chemotherapeutic that is administered orally, considered to have adequate and reliable absorption

It is usually well tolerated in small daily doses, although N/V can occur in a single daily dose >20mg

Clinically used to tx chronic lymphoblastic leukemia (CLL)

A

Chlorambucil

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

Describe nitrosureas as a class in terms of cross resistance with other alkylating agents, lipid solubility and related clinical implications, and chemical moieties generated

A

Not cross resistant with other alkylating agents

Lipid soluble — can cross the blood brain barrier making them effective at treating brain tumors

Generate both alkylating and carbamylating moieties

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

Major action and lipid solubility of carmustine

A

Major action is its alkylation of DNA — this can be repaired by MGMT

Highly lipophilic — crosses BBB

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

AEs of carmustine

A

Profound and delayed myelosuppression — recovery 4-6 weeks after a single dose

In high doses with bone marrow rescue, carmustine produces hepatic VOD, pulmonary fibrosis, renal failure, and secondary leukemia

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

Clinical applications of carmustine

A

Malignant gliomas (implantable carmustine wafer); [Methylation of the MGMT promoter inhibits its expression of 30% of primary gliomas]

Associated with sensitivity to carmustine and other nitrosoureas

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

Chemotherapeutic with high affinity for cells of islets of Langerhans, causing diabetes in experimental animals

Used clinically to tx human pancreatic islet cell carcinoma and carcinoid tumors

A

Streptozocin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

AEs of streptozocin

A

Frequent nausea

Mild reversible renal or hepatic toxicity occurring in approx 2/3 of cases (10% will have cumulative dose renal toxicity that can lead to renal failure)

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

Aziridine chemotherapeutic that is rapidly converted by hepatic CYPs to its desulfurated primary metabolite; used for high-dose chemotherapy regimens in transplants for hematological malignancies

A

Thiotepa (primary metabolite = TEPA)

[both thiotepa and TEPA from DNA cross links]

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

AEs of thiotepa

A

Little toxicity other than myelosuppression

AEs at high doses include mucosal and CNS toxicity, coma and seizures

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

Orally administered alkylsulfonate chemotherapeutic used to treat chronic myeloid leukemia

A

Busulfan

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

AEs of busulfan

A

At the standard dose — causes myelosuppression

At high doses — causes pulmonary fibrosis, GI mucosal damage, and hepatic VOD (veno-occlusive disease)

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

When a pt is receiving chemotherapy with busulfan, what drugs are given concomitantly and why?

A

Anti-convulsants — to protect against acute CNS toxicities

Phenytoin — increases GSTs that metabolize busulfan; reduces AUC by 20%

Non-enzyme inducing benzodiazepines recommended (lorazepam and clonazepam)

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

Non-classic alkylating agent with incompletely understood MOA; converted by CYP mediated oxidative metabolism to highly reactive alkylating species that methylate DNA; can produce chromosomal damage including chromatid breaks and translocations consistent with its mutagenic and carcinogenic actions, inhibits DNA, RNA, and protein biosynthesis, prolongs interphase (cell cycle), produces chromosome breaks

A

Procarbazine

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

Resistance to procarbazine develops rapidly when its used as a single agent (i.e., increased expression of MGMT). What are other AEs of procarbazine?

A

Carcinogenic potential higher than that of other alkylating agents

Increased risk of secondary cancers — acute leukemia

Augments sedative effects — concomitant CNS suppressants should be avoided

Disulfuram-like actions — avoid alcohol

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

Clinical applications of procarbazine

A

Used in the MOPP regimen for Hodgin disease

Also used in treating gliomas as a part of the PVC regimen (procarbazine + vincristine + lomustine [CCNU])

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

The prodrug of ______ functions as a methylating agent after being converted to monomethyl triazeno metabolite MTIC

Resistance develops due to __________

It is administered parenterally

A

Dacarbazine

Removal of methyl groups by MGMT

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

AEs of dacarbazine

A

Myelosuppression

N/V (90% of pts) — vomiting develops 1-3 hours after treatment and may last up to 12 hours; can be severe

Flu-like syndrome may occur

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

Clinical application of dacarbazine

A

Primary indication = Hodgkin disease

Modestly effective against malignant melanoma and adult sarcomas

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

MOA of bendamustine

A

Forms cross-links with DNA resulting in single and double stranded breaks —> inhibition of DNA synthesis and function

Inhibits mitotic check points and induces mitotic catastrophe —> cell death

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

Bendamustine’s cross resistance with other alkylating agents is only partial. What are adverse effects of Bendamustine?

A

Myelosuppression (rapidly reversible)

Mucositis (rapidly reversible)

Mild nausea and vomiting

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

Clinical applications of bendamustine

A

Approved for tx of CLL (chronic lymphoblastic leukemia) and non-Hodgkin lymphoma

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

MOA of the alkylating agents that are platinum analogs

A

[precise MOA unknown]

Kills tumor cells in all stages of the cell cycle

Binds DNA through formation of intra- and inter-strand cross links

Inhibits DNA synthesis and function

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

Platinum analog alkylating agents exhibit synergism with other alkylating agents, ______, and ______

A

Fluoropyrimidines; taxanes

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

Platinum analog only given by IV, requiring preceding tx with chloride diuresis via 1-2L NS to prevent renal toxicity, as well as vigorous IV hydration

A

Cisplatin

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

_____ inactivates cisplatin, thus the drug should not come into contact with needles or other infusion equiment containing it

A

Aluminum

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

T/F: cisplatin has abundant CNS penetration

A

False — it is found in highest concentrations in kidney, liver, intestine, and testes

It has poor CNS penetration

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

AEs of cisplatin

A

Nephrotoxicity [diminished by pretreatment with hydration and chloride diuresis]

Ototoxicity — manifests as tinnitus and leads to high frequency hearing loss

N/V occur in almost all pts

High doses/multiple tx may lead to progressive peripheral motor and sensory neuropathy (may worsen after discontinuation)

Mild to moderate disturbances in electrolytes — tubular damage may lead to renal electrolyte wasting and may produce tetany if untreated

Anaphylactic rxns may occur w/i minutes after administration (facial edema, bronchoconstriction, tachycardia, hypotension)

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

Treatment with cisplatin can lead to the development of what hematologic malignancy, usually 4+ years after treatment?

A

AML

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

How is the common adverse effect of N/V managed during cisplatin tx, as well as tx with other chemotherapy drugs?

A

5-HT3 receptor antagonists

NK1 receptor antagonists

High dose corticosteroids

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

Clinical applications for cisplatin therapy

A

Cisplatin in combo with bleomycin or etoposide (and with added vinblastine) cures 90% of pts with testicular cancer

Cisplatin used with paclitaxel induces complete response in majority of pts with ovarian carcinoma

Other cancers that respond to cisplatin: bladder, head and neck, cervix, endometrium, lung, rectal, anal, childhood neoplasms

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

______ has a similar MOA, resistance, and spectrum of activity as cisplatin but is a very different chemically, pharmacokinetically, and in terms of toxic properties. It is given IV and does NOT require vigorous IV hydration like cisplatin does

A

Carboplatin

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

AEs of carboplatin

A

Well tolerated clinically with less nausea, neurotoxicity, ototoxicity and nephrotoxicity than cisplatin

Dose limiting toxicity is myelosuppression —> thrombocytopenia

Hypersensitivity reaction — managed by administering graded doses of the drug and more prolonged infusions which lead to desensitization

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

Clinical applications of carboplatin

A

Carboplatin and cisplatin are equally effective in the tx of debulked ovarian cancer, non-small cell lung cancer, extensive-stage small cell lung cancer

Used with paclitaxel to induce complete response in majority of ovarian carcinoma

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

In terms of germ cell, head and neck, and esophageal cancers — which is more effective: carboplatin or cisplatin?

A

Cisplatin

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

Carboplatin can be an effective alternative to cisplatin in pts with what conditions?

A

Impaired renal function

Refractory nausea

Significant hearing impairment

Neuropathy

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

Oxaliplatin has the same mechanism as the other platinum analogs. What are some adverse effects of oxaliplatin?

A

Peripheral sensory neuropathy (acute form vs. cumulative dose)

Nausea is well controlled with 5-HT3 receptor antagonists

May cause leukemia and pulmonary fibrosis months to years after administration

May elicit an allergic response — urticaria, hypotension, and bronchoconstriction

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

Describe acute vs. cumulative dose forms of peripheral sensory neuropathy caused by oxaliplatin

A

Acute form: often triggered by exposure to cold liquids; paresthesias or dysesthesias in the upper and lower extremities, mouth, and throat

Cumulative dose: similar to cisplatin neuropathy; progressive sensory neurotoxicity — dysesthesias, ataxia, numbness of extremities

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

Clinical applications of oxaliplatin

A

Colorectal and gastric cancer

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

Oxaliplatin suppresses expression of _____ _____, which may be the rationale for the synergy with 5-FU. Oxaliplatin is approved in combination with 5-FU for _________ cancer

A

Thymidylate synthase (TS); colorectal

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

MOA of methotrexate

A

Folic acid analog that binds to the active site of dihydrofolate reductase

Inhibits synthesis of tetrahydrofolate (THF) [the one-carbon carrier for de novo synthesis of thymidylate, purine nucleotides, and amino acids serine and methionine]

Interference in formation of DNA, RNA, and key cell proteins

[Folyl polyglutamate synthase (FPGS) catalyzes formation of intracellular polyglutamate metabolites which are selectively retained in cancer cells]

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

4 Resistance mechanisms to methotrexate

A

Decreased drug transport via reduced folate carrier or folate receptor protein

Decreased formation of cytotoxic methotrexate polyglutamates

Increased levels of target enzyme DHFR through gene amplification and other genetic mechanisms

Altered DHFR protein with reduced affinity for methotrexate

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

Methotrexate may be administered IV, intrathecal, or orally. What 4 drugs inhibit the renal excretion of methotrexate, thus leading to increased risk of methotrexate toxicity?

A

Aspirin

NSAIDs

Penicillin

Cephalosporins

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

Methotrexate action can be reversed by administration of reduced folate ________, a drug sometimes used in conjunction with high dose methotrexate therapy to rescue normal cells from undue toxicity

A

Leucovorin

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

Clinical applications of methotrexate

A

Breast cancer

Head and neck cancer

Osteogenic sarcoma

Primary CNS lymphoma

Non-Hodgkins lymphoma

Bladder cancer

Choriocarcinoma

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

Antimetabolite chemotherapeutic active in the S-phase and transported into the cell via reduced folate carrier and requires activation by FPGS to yield higher polyglutamate forms (like methotrexate)

A

Pemetrexed

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

MOA of pemetrexed

A

Inhibition of thymidylate synthase (TS)

Targets and inhibits DHFR and enzymes involved in de novo purine nucleotide biosynthesis

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

Adverse effects of pemetrexed

A

Myelosuppression, skin rash, mucositis, diarrhea, fatigue, and hand-foot syndrome (painful erythema and swelling — tx with dexamethasone)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

Vitamin supplement with _____ and _____ significantly reduces toxicities associated with pemetrexed and pralatrexate

A

Folic acid and vitamin B12

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

Clinical applications of pemetrexed

A

Mesothelioma

Approved for use in combo with cisplatin for the first-line tx of NSCLC and as maintenance therapy in pts with NSCLC whose disease has not progressed after 4 cycles of platinum-based chemotherapy

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

Antimetabolite similar to methotrexate in that it is transported into the cell via reduced folate carrier (RFC) and requires activation by FPGS to yield higher polyglutamate forms, but designed to be more potent substrate for the RFC and FPGS

A

Pralatrexate

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

MOA of pralatrexate

A

Inhibits DHFR

Inhibits enzymes involved in de novo purine nucleotide biosynthesis

Inhibits thymidylate synthase

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

AEs of pralatrexate

A

Myelosuppression, skin rash, mucositis, diarrhea, fatigue

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

Clinical application for pralatrexate

A

Approved for tx of relapsed or refractory peripheral T-cell lymphoma

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

Active metabolites of 5-FU and their MOA

A

FdUMP — binds and forms a ternary complex with TS and reduced 5,10-methylenetetrahydrofolate; inhibition of DNA synthesis through “thymineless death”

FUTP — incorporated into RNA where it interferes with RNA processing and mRNA translation

FdUTP — incorporated into DNA resulting in inhibition of DNA synthesis and function

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

5-FU is administered IV and has an extremely _____ half life; it is catabolized by the enzyme ___________

A

Short (10-15 mins)

Dihydropyrimidine dehydrogenase (DPD)

[AR syndrome associated with partial or complete deficiency of the DPD gene seen in 5% of pts — leads to toxicities of myelosuppresion, GI toxicity with diarrhea/mucositis, and neurotoxicity]

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

AEs of 5-FU

A

Myelosuppression

GI toxicity (diarrhea/mucositis)

Skin toxicity (hand-foot syndrome)

Neurotoxicity

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

Clinical applications of 5-FU

A

Activity against a wide range of solid tumors — breast, stomach, pancreas, esophagus, liver (hepatocellular), head and neck, and anus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

Most widely used drug in the tx of colorectal cancer (both as an adjuvant and for advanced disease)

A

5-FU

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

Prodrug metabolized to 5-FU in the cancer cell by thymidine phosphorylase, at which time MOA is the same as 5-FU

A

Capecitabine

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

The expression of thymidine phosphorylase is higher in what type of tumors (as opposed to normal tissue)?

A

Solid tumors (e.g., breast, colorectal cancer)

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

AEs of capecitabine

A

Main AEs = diarrhea, hand-foot syndrome

Others that occur but less frequently than with 5-FU: myelosuppression, N/V, mucositis, and alopecia

89
Q

Clinical applications of capecitabine

A

Metastatic breast cancer as single agent, or combined with others (docetaxel, paclitaxel, lapatinib, ixabepilone, and trastuzumab)

Approved for use in adjuvant therapy of stage III and high risk stage II colon cancer

Metastatic colorectal cancer in combo with irinotecan and oxaliplatin

90
Q

The XELOX regimen is considered first line tx for metastatic colorectal cancer. What is included in this regimen?

A

Capecitabine + oxaliplatin

[also used in adjuvant setting for stage III and high-risk stage II colon cancer]

91
Q

TAS-102 is a prodrug that is inactive in its parent form; an antimetabolite approved in 2015. What are its components?

A

Trifluridine/tipiracil (1:0.5 ratio)

Trifluridine is a fluorinated pyrimidine nucleoside analog that is metabolized to the monophosphate form — inhibits thymidylate synthase (TS) although much weaker than FdUMP; when metabolized into the triphosphate form, it is directly incorporated into DNA and inhibits its synthesis and function

Tipiracil is a thymidine phosphorylase (TP) inhibitor [key enzyme in degrading trifluridine to inactive forms] which leads to higher levels of trifluridine

92
Q

Clinical applications for TAS-102

A

Approved for use in progressive, refractory colorectal cancer

[retains clinical activity in the setting of wild-type and mutant RAS colorectal cancer]

93
Q

Dose-limiting toxicity and other adverse effects of TAS-102

A

Myelosuppression (neutropenia more commonly observed than anemia and thrombocytopenia)

Other AEs: GI toxicity with diarrhea and N/V, fatigue, anorexia

94
Q

S-phase specific antimetabolite converted to the 5’-mononucleotide (ara-CMP) by deoxycytidine kinase which is further metabolized to diphosphate and triphosphate metabolites

A

Cytarabine

[the triphosphate metabolite (ara-CTP) is thought to be main cytotoxic metabolite]

95
Q

MOA of cytarabine

A

Ara-CTP competitively inhibits DNA polymerase-alpha (blockade of DNA synthesis) and DNA polymerase-beta (blockade of DNA repair)

Ara-CTP also incorporated into DNA —> interference with chain elongation; defective ligation of fragments of newly synthesized DNA

Ara-CTP also incorporated into RNA

96
Q

An important consideration regarding cytarabine is that it is rapidly degraded, but cellular retention of ara-CTP correlates with its lethality to malignant cells — so it is often given by continuous infusion. What are some adverse effects of cytarabine therapy?

A

Myelosuppression
Mucositis
N/V
Neurotoxicity (high dose)

97
Q

Clinical applications of cytarabine

A

Activity is limited to hematologic malignancies, including AML and NHL

[No activity against solid tumors!]

98
Q

Fluorine-substituted deoxycytidine analog that is phosphorylated by deoxycytidine kinase to the monophosphate form, or by other nucleoside kinases to diphosphate and triphosphate nucleotide forms

A

Gemcitabine

99
Q

MOA of gemcitabine diphosphate and triphosphate metabolites

A

Gemcitabine diphosphate — inhibition of ribonucleotide reductase (reduces level of deoxyribonucleoside triphosphates needed for DNA synthesis)

Gemcitabine triphosphate — inhibits DNA polymerase-alpha (blockade of DNA synthesis) and DNA polymerase-beta (blockade of DNA repair); incorporates into DNA leading to inhibition of DNA synthesis and function

100
Q

Gemcitabine has dose-limiting toxicity of myelosuppression in the form of neutropenia. It also has AEs of N/V (70% of pts) and a flu-like syndrome. What are some clinical applications of gemcitabine?

A

Broad spectrum activity against both solid tumors and hematologic malignancies

Initially approved for use in advanced pancreatic cancer

Widely used to treat many different cancers — NSCLC, bladder cancer, ovarian cancer, soft tissue sarcoma, NHL

101
Q

6-thiopurines are metabolized by the enzyme _____ ______, thus loss of this enzyme alters the metabolism of thiopurines and can result in increased toxic effects including myelosuppression and GI toxicity with mucositis and diarrhea

A

Thiopurine methyltransferase (TPMT)

102
Q

6-mercaptopurine (6-MP) is a purine antagonist inactive in its parent form, metabolized hy hypoxanthine-guanine phosphoribosyl transferase (HGPRT) to form monophosphate nucleotide 6-thioinosinic acid. What is the MOA of the active metabolites?

A

Monophosphate nucleotide 6-thioinosinic acid inhibits several enzymes of de novo purine nucleotide synthesis

Triphosphate form incorporated into both DNA and RNA

103
Q

6-MP is converted to its inactive metabolite (6-thiouric acid) by an oxidation reaction catalyzed by _____ _____.

Thus, the purine analog _______, a potent inhibitor of the above enzyme, is frequently used as a supportive care measure in the tx of acute leukemias to prevent the development of hyperuricemia that often occurs after tumor cell lysis

A

Xanthine oxidase

Allopurinol

[note that simultaneous therapy with allopurinol and 6-MP would result in increased levels of 6-MP and excessive toxicity]

104
Q

Single clinical application of 6-MP

A

Childhood acute leukemia

105
Q

MOA of 6-thioguanine (6-TG)

A

Inhibits several enzymes in the de novo purine nucleotide biosynthetic pathway

Inhibition of purine nucleotide interconversion

Decrease in intracellular levels of guanine nucleotides —> inhibition of glycoprotein synthesis

Interference with the formation of DNA and RNA

Incorporation of thiopurine nucleotides into both DNA and RNA

106
Q

6-TG synergizes with ______ in the treatment of adult acute leukemia. It is converted to an inactive metabolite by the process of _______

A

Cytatabine; deamination

107
Q

Fludarabine is a purine antagonist that is phosphorylated intracellularly by deoxycytidine kinase to the monophosphate and eventually converted to the triphosphate form. What is the MOA of these metabolites?

A

Fludarabine triphosphate — interferes with process of DNA synthesis and DNA repair through inhibition of DNA polymerase-alpha and beta. Directly incorporated into DNA, resulting in inhibition of DNA synthesis and function

Fludarabine diphosphate — inhibits ribonucleotide reductase leading to an inhibition of the production of essential deoxyribonucleotide triphosphates

Induces apoptosis in susceptible cells through a yet to be determined mechanism of action

108
Q

Dose limiting toxicity and other AEs of fludarabine

A

Dose limiting = myelosuppression

AEs:
Potent immunosuppressant —> increased risk for opportunistic infections; pneumocystis jiroveci pneumonia (PCP)

[PCP prophylaxis with TMP/SMX at least 3x/week; prophylaxis should continue up to 1 week after stopping fludarabine tx]

109
Q

Clinical applications of fludarabine

A

Low-grade NHL

Chronic lymphocytic leukemia

110
Q

Purine nucleoside analog with high specificity for lymphoid cells; inactive in its parent form — initially phosphorylated by deoxycytidine kinase to the monophosphate form and eventually metabolized to the triphosphate form

A

Cladribine

111
Q

MOA of cladribine triphosphate metabolite

A

Directly incorporated into DNA, resulting in inhibtion of DNA synthesis and function; interferes with the process of DNA synthesis and repair through inhibition of DNA polymerase alpha and beta

112
Q

The main toxicity of cladribine is _______.

Cladribine is administered as a continuous 7-day infusion, and there is very manageable side effect profile under these conditions

However, a decrease in ____ and _____ cells can last over 1 year

A

Myelosuppression

CD4; CD8

113
Q

Clinical applications for cladribine

A

Hairy cell leukemia

Other low-grade lymphoid malignancies — CLL and NHL

114
Q

MOA of vinca alkaloids class of chemotherapeutics

A

Inhibit tubulin polymerization, disrupting assembly of microtubules (especially those involved in mitotic spindle, leading to mitotic arrest in metaphase, leading to cell death)

M-phase specific!

Microtubules also have important roles in maintaining cell shape and protein transport, so disruptions in these processes can lead to cell death as well

115
Q

Vinca alkaloid derived from periwinkle plant Vinca rosea, used in Hodgkins and non-hodgkins lymphomas, breast cancer, and germ cell cancers

A

Vinblastine

116
Q

AEs of vinblastine

A

N/V
Bone marrow suppression
Alopecia
Potent vesicant (blistering)

117
Q

Vinca alkaloid derived from the periwinkle plant Vinca rosa. Clinical uses include pediatric acute lymphoblastic leukemia (in combo with prednisone), Hodgkins and non-hodgkins lymphoma, multiple myeloma, and pediatric tumors including rhabdomyosarcoma, neuroblastoma, Ewing’s sarcoma, and Wilm’s tumor

A

Vincristine

[Vincristine is closely related structurally to vinblastine with similar MOA, mech of resistance, and clinical pharmacology, but different in terms of clinical activity and safety profile]

118
Q

Vincristine has a higher affinity for _____ microtubules

A

Axonal

119
Q

Dose limiting toxicity of vincristine

A

Neurotoxicity (usually peripheral sensory neuropathy)

— can be ANS dysfunction with orthostatic hypotension, urinary retention, paralytic ileus or constipation, cranial nerve palsies, ataxia, seizures, coma

120
Q

Aside from its dose-limiting toxicity of neurotoxicity, what are some other AEs of vincristine?

A

Myelosuppression (although milder and less significant that vinblastine)

SIADH

121
Q

Semisynthetic derivative of vinblastine with same MOA as vinblastine and vincristine; used in NSCLC, breast cancer, and ovarian cancer

A

Vinorelbine

122
Q

Dose limiting toxicity and AEs associated with vinorelbine

A

Dose limiting: Myelosuppression with neutropenia

AEs: N/V, transient elevations in LFTs, neurotoxicity, and SIADH

123
Q

Natural products like the taxanes and other anti-microbial drugs work in the ____ phase of the cell cycle

A

M phase

124
Q

Alkaloid ester derived from the Pacific yew (Taxus brevifolia) and the European yew (Taxus baccata) that acts as a mitotic spindle poison, binding with high affinity to microtubules with enhancement of tubulin polymerization, leading to inhibition of mitosis and cell division

A

Paclitaxel

125
Q

Acute and delayed dose-limiting toxicities associated with paclitaxel

A

Acute — N/V, hypotension, arrhythmias, hypersensitivity

Delayed — myelosuppression, peripheral sensory neuropathy

126
Q

Hypersensitivity reactions to paclitaxel are observed in 5% of pts. The incidence of these are significantly reduced by premedication with _____, _____, AND _______

A

Dexamethasone, diphenhydramine, H2 blocker

127
Q

What are the benefits of the albumin-bound paclitaxel nanoparticle formulation?

A

Not associated with hypersensitivity reactions

Reduced myelosuppressive effects compared with paclitaxel

Neurotoxicity is more readily reversed when compared with paclitaxel

128
Q

Clinical applications of paclitaxel

A

Broad range of solid tumors:

Ovarian, advanced breast, NSCLS and small cell lung cancer

Head and neck, esophageal, prostate, and bladder

AIDS-related Kaposi’s sarcoma

129
Q

Semisynthetic taxane derived from the European yew tree with identical MOA, metabolism, and elimination to paclitaxel; approved as second line therapy in advanced breast cancer and NSCLC, with major activity in head and neck cancer, small cell lung cancer, gastric cancer, advanced platinum-refractory ovarian cancer, and bladder cancer

A

Docetaxel

130
Q

Acute vs. delayed toxicities of docetaxel

A

Acute: hypersensitivity

Delayed: neurotoxicity, fluid retention, myelosuppression with neutropenia

131
Q

Semisynthetic taxane with identical MOA, metabolism, and elimination to other taxanes, but a unique characteristic is that it is a poor substrate for P-glycoprotein, making it useful for treating multi-drug resistant tumors

A

Cabazitaxel

132
Q

Major toxicities of cabazitaxel

A

Myelosuppression
Neurotoxicity
Allergic reactions

133
Q

Semisynthetic epothilone B analog (NOT a taxane) that acts as a microtubule inhibitor, binding directly to beta-tubulin subunits leading to dynamic inhibition of normal microtubule function

A

Ixabepilone

134
Q

Ixabepilone is active in the M phase of the cell cycle and has activity in resistant tumors with _____ mutations and overexpression of __________

A

Tubulin; P-glycoproteins

135
Q

AEs of ixabepalone

A

Myelosuppression

Hypersensitivity

Neurotoxicity (peripheral sensory neuropathy)

136
Q

Clinical applications of ixabepilone

A

Approved for metastatic breast cancer (as a monotherapy or in combo with capecitabine)

137
Q

Synthetic analog of halichondrin B, works to inhibit microtubule function leading to block in the G2-M phase of the cell cycle

A

Eribulin

138
Q

Eribulin is less sensitive to P-glycoprotein, so it has activity in tumors overexpressing P-glycoprotein. It is approved for what type of cancer?

A

Metastatic breast cancer

139
Q

Semisynthetic derivative of podophyllotoxin, extracted from mayapple root (Podophyllum peltatum); available in IV and oral formulations for the tx of germ cell cancer, small cell and NSCLC, Hodgkins and nonhodgkins lymphoma, and gastric cancer

A

Etoposide

140
Q

MOA of etoposide

A

Forms a complex with topoisomerase II and DNA, leading to inhibition of the activity of topoisomerase II and subsequent inactivation of DNA synthesis and function

141
Q

Major acute vs. delayed toxicities of etoposide

A

Acute: N/V, hypotension

Delayed: alopecia, myelosuppression

142
Q

Natural Chinese tree-derived class of chemotherapeutics that work by inhibiting the activity of topoisomerase I, resulting in DNA damage

A

Camptothecins (topotecan and irinotecan)

143
Q

AEs of camptothecans (topotecan and irinotecan)

A

Myelosuppression and diarrhea

[early form of diarrhea occur within 24 hrs of administration as cholinergic event; may be treated with atropine. Late form of diarrhea occurs 2-10 days later and can be severe — leading to electrolyte imbalances and dehydration]

144
Q

IV administered camptothecan analog that is indicated for treatment of advanced ovarian cancer as second-line therapy (following initial tx with platinum-based chemotherapy) as well as second-line therapy for small cell lung cancer

A

Topotecan

145
Q

Dose limiting toxicity of topotecan

A

Neutropenia

146
Q

Irinotecan is a prodrug camptothecan analog, converted by carboxylesterase enzyme to ______ in the ______, a metabolite that is a 1000x more potent topoisomerase I inhibitor vs irinotecan

A

SN-38; liver

147
Q

Clinical applications of irinotecan

A

Approved as first-line therapy when used in combo with 5-FU and leucovorin for metastatic colorectal cancer

[Originally approved as second-line monotherapy for metastatic colorectal cancer]

148
Q

Little is known about the clinical pharmacology or metabolism of liposomal irinotecan, but it is approved in combination with 5-FU and leucovorin for the treatment of ___________ after disease progression following gemcitabine-based therapy

A

Adenocarcinoma of the pancreas

[AEs of liposomal irinotecan include myelosuppression and GI toxicity with diarrhea, N/V]

149
Q

The anthracyclines are anti-tumor antibiotics isolated from Streptomyces peucetius var caesius and are among the most widely used cytotoxic anti-cancer drugs. What is the MOA of anthracyclines?

A

Inhibition of topoisomerase II

Generation of semiquinone free radicles and oxygen free radicals through iron-dependent, enzyme-mediated reductive process

High affinity binding to DNA through intercalation, with blockade of the synthesis of DNA and RNA, and DNA strand scission

Binding to cell membranes to alter fluidity and ion transport

150
Q

Part of the MOA of anthracyclines is the generation of semiquinone and oxygen free radicals, this has been well established as the cause of what toxicity associated with this drug class?

A

Cardiotoxicity

151
Q

Dose limiting toxicity and other AEs of anthracyclines

A

Dose-limiting = myelosuppression (neutropenia more common than thrombocytopenia); in some cases dose-limiting factor may be mucositis

Other AEs: cardiotoxicity, radiation recall reaction (erythema and desquamation of the skin observed at sites of prior radiation therapy)

152
Q

Acute vs. chronic form of cardiotoxicity seen with the anthracyclines

A

Acute: occurs w/i first 2-3 days. Presents with arrhythmias and conduction abnormalities, other ECG changes, pericarditis, and myocarditis. Transient and often asymptomatic.

Chronic: dose-dependent; dilated cardiomyopathy associated with heart failure, increased production of oxygen free radical production within the myocardium — IMPORTANT: lower the weekly doses or continue infusion to reduce likelihood of this toxicity

153
Q

_______ is an anthracycline generally combined with cyclophosphamide, cisplatin, and/or 5-FU, as it has greater activity in combination vs. monotherapy

A

Doxorubicin

154
Q

Doxorubicin is one of the most important cancer drugs in clinical practice, what are the clinical applications of doxorubicin?

A

Major clinical activity in cancers of the breast, endometrium, ovary, testicle, thyroid, stomach, bladder, liver, and lung [also soft tissue sarcomas, as well as childhood cancers including neuroblastoma, Ewing’s sarcoma, osteosarcoma, and rhabdomyosarcoma]

Clinical activity in hematologic malignancies: ALL, multiple myeloma, Hodgkins and non-hodgkins lymphomas

155
Q

______ is the first agent of the anthracycline class to be isolated, and unlike doxorubicin, its efficacy is limited in solid tumors. It is primarily used in tx of AML

A

Daunorubicin

156
Q

Semisynthetic antracycline glycoside analog of daunorubicin used in combo with cytarabine for induction therapy of AML

A

Idarubicin

[when combined with cytarabine, idarubicin is more active than daunorubicin in producing complete remissions and in improving survival in pts with acute myelogenous leukemia]

157
Q

Anthracycline analog whose mechanism of action and clinical pharm are identical to the others in this class; initially approved for use as a component of adjuvant therapy in early stage, node-positive breast cancer. Also used in tx of metastatic breast cancer and gastroesophageal cancer

A

Epirubicin

158
Q

Anthracene compound whose structure resembles the anthracycline ring; works by binding DNA to produce strand breakage and inhibits both DNA and RNA synthesis

A

Mitoxantrone

159
Q

Dose limiting and other toxicities associated with mitoxantrone

A

Dose limiting = myelosuppresion with leukopenia

AEs: mild nausea and vomiting, mucositis, and alopecia

Acute and chronic cardiac toxicity (although less so than docorubicin)

Blue discoloration of the fingernails, sclera, and urine is observed 1-2 days after drug administration

160
Q

Clinical application of mitoxantrone

A

Currently used in tx of advanced, hormone-refractory prostate cancer and low-grade-non-Hodgkin’s lymphoma

Indicated in breast cancer and in pediatric and adult acute myeloid leukemias

161
Q

Iron-chelating agent approved to prevent or reduce anthracycline-induced cardiotoxicity in women with metastatic breast cancer that have received a cumulative dose (>300 mg/m^2) of doxorubicin

A

Dexrazoxane

162
Q

Anti-tumor antibiotic isolated from streptomyces caespitosus; metabolically activated via enzyme-mediated reduction, generating an alkylating agent that cross-links DNA (works similar to other alkylating agents)

A

Mitomycin (mitomycin C)

163
Q

T/F: Inner solid tumor cells are more sensitive to mitomycin vs. normal cells

A

True — d/t hypoxic environment conducive to reductive reactions. The reduction of mitomycin to the alkylating agent is more likely to occur within the tumor

164
Q

Best available drug for use in combination with radiation therapy to attack hypoxic tumor cells

A

Mitomycin (mitomycin C)

165
Q

In what phase(s) of the cell cycle is mitomycin active?

A

All phases

166
Q

Acute and delayed toxicities associated with mitomycin

A

Acute: N/V

Delayed: Myelosuppression, mucositis, anorexia and fatigue, hemolytic uremic syndrome

167
Q

Clinical applications of mitomycin

A

Squamous cell cancer of the anus in combo with 5-FU and radiation

Intravesical tx of superficial bladder cancer (little systemic absorption of mitomycin when it is used in this way)

168
Q

Small peptide anti-tumor antibiotic that contains a DNA binding region and an iron-binding domain at opposite ends of the molecule; clinically indicated for Hodgkin’s and non-hodgkins lymphomas, germ cell tumors, head and neck cancer, squamous cell cancer of the skin, cervix, and vulva

A

Bleomycin

169
Q

MOA of bleomycin

A

Binds to DNA resulting in single and double strand breaks following free radical formation

Binds to DNA and inhibits DNA biosynthesis

DNA fragmentation is due to oxidation of a DNA-bleomycin-Fe(II) complex —> chromosomal aberrations

170
Q

The MOA of bleomycin causes accumulation of cells in the ____ phase of the cycle

A

G2

171
Q

Dose-limiting toxicity and other AEs associated with bleomycin

A

Dose-limiting: pulmonary toxicity (pneumonitis w/ cough, dyspnea, dry inspiratory crackles, infiltrates on CXR)

AEs: acute include allergic reactions and hypotension; delayed include skin toxicity, pulmonary fibrosis, mucositis, and alopecia

172
Q

In rare cases, the pulmonary toxicity that occurs with bleomycin therapy may be fatal. What factors increase the risk of experiencing pulmonary toxicity on bleomycin therapy?

A

Age >70

Cumulative dose >400 units

Underlying pulmonary disease

Prior mediastinal or chest irradiation

173
Q

Describe metabolism of the tyrosine kinase inhibitors and the drug and dietary implications of this

A

TK inhibitors are metabolized by CYP3A4 in the liver

Drug-drug interactions occur with medications that are also elminated by this route

Pts on these drugs should avoid grapfruit products, starfruit, and pomelos (which may inhibit metabolism of the TK inhibitors)

174
Q

TK inhibitor that works as inhibitor of Bcr-Abl oncoprotein, preventing phosphorylation of the kinase substrate by ATP; also inhibits other RTKs for platelet-derived growth factor receptor (PDGFR) and c-kit

A

Imatinib

175
Q

Acute and delayed toxicities associated with imatinib

A

Acute: N/V

Delayed: fluid retention with ankle and periorbital edema, diarrhea, myalgias, CHF

176
Q

Clinical applications for imatinib

A

CML (philadelphia chromosomal translocation t9:22)

GI stromal tumors expressing the c-kit tyrosine kinase

177
Q

First line therapy in chronic phase CML, in blast crisis, and as second-line therapy for chronic phase CML that has progressed on prior IFN-alpha therapy

A

Imatinib

178
Q

Inhibitor of several tyrosine kinases including Bcr-Abl, c-kit, and PDGFR-beta; differs from imatinib in that it binds to the active and inactive conformations of the Abl kinase domain (thus it can overcome imatinib resistance resulting from mutations in Bcr-Abl)

A

Dasatinib

[thus clinical applications of dasatinib are CML and philadelphia chromosome positive ALL with resistance or intolerance to imatinib therapy]

179
Q

Second generation phenylamino-pyrimidine molecule that inhibits Bcr-Abl, c-kit, and PDGFR-beta tyrosine kinases; differs from imatinib with higher affinity for the Abl kinase (thus overcoming imatinib resistance resulting from Bcr-Abl mutations)

A

Nilotinib

[thus main clinical use is chronic phase and accelerated phase CML with resistance or intolerance to prior therapy including imantinib; Also recently approved as first-line therapy in chronic phase CML]

180
Q

Inhibitor of Bcr-Abl tyrosine kinase that retains activity in 16 of the 18 imatinib-resistant Bcr-Abl mutations, but is not effective against T315I and V299L mutations which reside in the ATP-binding domain of the Abl tyrosine kinase

A

Bosutinib

181
Q

AEs of bosutinib

A

Acute: N/V

Delayed: diarrhea, fluid retention, myelosuppression, skin rash, hepatotoxicity

182
Q

Clinical applications of bosutinib

A

Adult pts with chronic, accelerated, or blast phase Ph chromosome-positive CML with resistance or intolerance to prior therapy

183
Q

Inhibitor of all known mutant forms of Bcr-Abl (including gate keeper mutation T315l) tyrosine kinase as well as other kinases including VEGF-R, PDGF, FGF, Flt3, TIE-2, Src family kinases, Kit, TET, and EPH

A

Ponatinib

184
Q

FDA approved clinical application of Ponatinib

A

Adult pts with chronic, accelerated, or blast phase CML that is resistant or intolerant to prior TKI therapy and also approved for Ph+ ALL that is resistant or intolerant to prior TKI therapy

185
Q

_____ is a receptor overexpressed in a number of solid tumors. Its activation and downstream signaling lead to cell growth and proliferation, invasion and metastasis, and angiogenesis. Signaling at this receptor inhibits the cytotoxic activity of various anti-cancer agents and radiation therapy leading to suppression of key apoptotic mechanisms and the development of cellular drug resistance.

A

Epidermal growth factor receptor (EGFR)

186
Q

VEGF is an angiogenic growth factor whose signaling pathway is an attractive target for chemotherapy. What are some general methods to inhibit VEGF signaling?

A

Inhibition of interaction of VEGF with its receptor — target VEGF ligand w/ Abs, compete with VEGF receptors for VEGF w/soluble chimeric decoy receptors

Direct inhibition of VEGF receptor-associated tyrosine kinase activity by small molecule inhibitors

187
Q

Chimeric monoclonal Ab directed against the extracellular domain of EGFR; used in combination with irinotecan and G1 isotype has antitumor activity mediated in part by immunologic-mediated mechanisms

A

Cetuximab

188
Q

The efficacy of cetuximab is restricted to those pts whose tumors express the wild-type _____ gene

A

RAS (includes both KRAS and NRAS)

189
Q

Adverse effects of cetuximab

A

Generally well tolerated

Acneiform skin rash

Hypersensitivity infusion reaction

Hypomagnesemia

190
Q

Clinical applications of cetuximab

A

Combined with irinotecan for metastatic colon cancer in the refractory setting or as monotherapy in pts who are deemed to be irinotecan-refractory

Combined with irinotecan+oxaliplatin in first line tx of metastatic colorectal cancer

Approved for use in combo with radiation therapy in pts with locally advanced head and neck cancer (BLACK BOX WARNING of increased risk of sudden death when combined with radiation for head/neck cancer)

191
Q

Fully human monoclonal antibody directed against EGFR, MOA is inhibition of EGFR signaling pathway but the G2 isotype of this drug is not expected to exert any immunologic-mediated effects

A

Panitumumab

192
Q

The efficacy of panitumumab is restricted to those patients whose tumors express the wild-type ___ gene. Panitumumab can be combined with _____

A

RAS; irinotecan

193
Q

Adverse effects of panitumumab

A

Acneiform skin rash and hypomagnesemia

Infusion related reactions can occur but are less common than cetuximab bc panitumumab is fully human antibody

194
Q

Clinical applications for panitumumab

A

Originally approved for pts with refractory metastatic CRC who have been tx with all other active agents

Approved for use with the FOLFOX chemotherapy regimen in the front-line treatment of metastatic CRC [FOLFOX = oxaliplatin + 5-FU + folinic acid (leucovorin)]

Combined with irinotecan-based therapy in second-line tx of metastatic CRC

195
Q

Fully human IgG1 monoclonal Ab directed against EGFR; the G1 isotype of this drug has antitumor activity mediated in part by immunologic-mediated mechanisms

A

Necitumumab

196
Q

Necitumumab has the same adverse effects as cetuximab and panitumab with the addition of increased risk of _____ and _____ events

A

Venothrombolic; arterioembolic

197
Q

Clinical application for necitumumab

A

Approved for use in combo with gemcitabine and cisplatin chemotherapy for the tx of squamous NSCLC

198
Q

Small molecule inhibitor of the tyrosine kinase domain associated with EGFR, metabolized by CYP3A4 (potential for drug interactions with things like phenytoin and warfarin; grapefruit products)

A

Erlotinib

199
Q

Erlotinib is often combined with _______

Patients who are ______ are more responsive to erlotinib

A

Gemcitabine

Nonsmokers

200
Q

AEs of erlotinib include acneiform rash, diarrhea, anorexia, and fatigue. What are its clinical applications?

A

First-line tx of metastatic NSCLC in pts with EGFR mutations and in pts that are refractory to at least one prior chemotherapy regimen

Maintenance therapy of pts with metastatic NSCLC whose disease has not progressed after 4 cycles of platinum-based chemotherapy

Approved for use in combo with gemcitabine for the treatment of advanced pancreatic cancer

201
Q

Small molecule inhibitor of the tyrosine kinase domains associated with EGFR, HER2, and HER4 —> inhibits downstream ErbB signaling

A

Afatinib

202
Q

Afatinib has similar toxicities to erlotinib (acneiform rash, diarrhea, anorexia, fatigue); what is its clinical application?

A

First-line tx of metastatic NSCLC with EGFR mutations

203
Q

Small molecule inhibitor that treats metastatic EGFR T790M mutant NSCLC following progression on or after EGFR tyrosine kinase inhibitor therapy; also targets the L858R and exon 19 EGFR mutations

A

Osimertinib

204
Q

Osimertinib has similar AEs to erlotinib and afatinib in addition to what unique toxicity?

A

Cardiotoxicity — QTc prolongation and cardiomyopathy

205
Q

Recombinant humanized monoclonal antibody that targets all forms of VEGF-A by binding to it and preventing interaction with target receptors

A

Bevacizumab

206
Q

Bevacizumab does not exacerbate the toxicities typically observed with cytotoxic chemo; what are th emain concerning AEs with bevacizumab?

A

HTN

Increased incidence of arterial thromboembolic events (TIA, stroke, angina, MI)

Wound healing complications

GI perforation

Proteinuria

207
Q

Clinical applications for bevacizumab

A

Combined with 5-FU and oxaliplatin in tx of metastatic CRC

FDA approved as first-line treatment for metastatic CRC in combo with any IV fluoropyrimidine-containing regimen

Also approved in combo with chemo for metastatic NSCLC and breast cancer

208
Q

Recombinant fusion protein made up of portions of the extracellular domains of human VEGF receptors 1 and 2 fused to the Fc portion of the human IgG1 molecule (soluble receptor to VEGF-A, VEGF-B, and placental growth factor [PIGF]), binds VEGF-A with significantly higher affinity vs. bevacizumab

A

Ziv-aflibercept

209
Q

Ziv-aflibercept has similar adverse effects as bevacizumab. What are its clinical applications?

A

Used in combo with FOLFIRI regimen for pts with metastatic CRC that has progressed on oxaliplatin-based chemotherapy

210
Q

IgG1 antibody that targets the VEGF-R2 receptor and inhibits the binding of VEGF-A, VEGF-C, and VEGF-D, thus inhibiting downstream VEGF-R2 signaling

A

Ramucirumab

211
Q

Ramucirumab has similar AEs as bevacizumab. What are the clinical applications of ramucirumab?

A

Advanced gastric or gastroesophageal junction adenocarcinoma, metastatic NSCLC, and metastatic CRC

212
Q

Small molecule that inhibits multiple RTKs and is metabolized by the CYP3A4 system

A

Sorafenib

[inhibits VEGF-R2, VEGF-R3, platelet-derived growth factor beta, and raf kinase]

213
Q

AEs of sorafenib

A

HTN

Bleeding complications

Fatigue

Skin rash and hand-foot syndrome observed in 30-50%

214
Q

Clinical applications for sorafenib

A

Initially approved for renal cell cancer

Also approved for advanced hepatocellular cancer

215
Q

Similar drug to sorafenib that inhibits multiple RTKs including PDGFR-alpha and beta, VEGF-R1/R2/R3, and c-kit; may be utilized as an alternative to imatinib after tolerance has developed

A

Sunitinib

216
Q

Sunitinib is metabolized by CYP3A4 system, so drug interactions must be monitored for. What are the main complications associated with Sunitinib?

A

HTN

Bleeding complications

Fatigue

Increased risk of cardiac dysfunction — can lead to CHF

217
Q

Clinical applications for sunitinib

A

Advanced renal cell cancer

GI stromal tumors after disease progression on or with intolerance to imatinib

218
Q

Small molecule that inhibits multiple RTKs including VEGF-R2/R3, PDGFR-beta, and raf kinase; metabolized by CYP3A4 system, and is only approved for the tx of advanced renal cell cancer

A

Pazopanib

[adverse effects include HTN, bleeding complications, and fatigue similar to others in this class]