Onco drugs Flashcards

1
Q

Cyclophosphamide

A

alkylating agent

Cross-link DNA at guanine
N-7. Require bioactivation by liver. A nitrogen mustard.

USE: Solid tumors, leukemia, lymphomas.

ADVERSE EFFECTS: Myelosuppression; hemorrhagic
cystitis, prevented with
mesna (thiol group of mesna binds toxic metabolites) or N-acetylcysteine.

Mechanism of Action - Forms DNA cross-links, resulting in inhibition of DNA synthesis & function

Toxicity - Acute: (GI)-Nausea & Vomiting
- Delayed: Moderate depression of peripheral blood count {Excessive doses lead to severe bone marrow depression with leukopenia, thrombocytopenia, and bleeding (hemorrhagic cystitis)}

Clinical Applications -
Breast and (Ovarian cancers - Hematologic Malignancies: NHL & CLL - Soft tissue sarcoma - Neuroblastoma - Wilms Tumor - Rhabdomyosarcoma

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

Procarbazine

A

Alkylating agent, non classic

Mechanism of action
– Inhibits DNA, RNA, & protein synthesis • Toxicity
– Acute: Nausea and Vomiting
– Delayed: Myelosuppression, CNS toxicity (when used with MAO inhibitors),
– **High risk of secondary malignancy

Oral agent – Cytotoxic drug metabolite acts as Monoamine
Oxidase inhibitor (MAOI) – Adverse drug-drug interactions with:
• Alcohol
• Sympathomimetic amines
• Tricyclic antidepressants
• Anti-histamines
• CNS depressants

• Clinical Applications
– Hodgkin’s lymphoma
– Non-Hodgkins lymphoma
– Brain tumors
– Component of MOPP** regimen
    **MOPP- Mechlorethamine, vincristine (Oncovorin®), Procarbazine, Prednisone
Rx: Hodgkins Disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Platinum based, alkylating agents

A

• Mechanism of Action
– “stick” to the DN! strands
– **Cytotoxic in ALL stages of cell cycle •

Metabolism: **Renal excretion
• Bind cytoplasmic & nuclear proteins too
• Usually used in combination. Synergize with:
– Other alkylating agents
– Fluoropyrimidines
– Taxanes

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

Cisplatin

A

Cisplatin, carboplatin
MECHANISM: Cross-link DNA.
CLINICAL USE Testicular, bladder, ovary, and lung carcinomas.
ADVERSE EFFECTS Nephrotoxicity, peripheral neuropathy, ototoxicity. Prevent nephrotoxicity with amifostine (free
radical scavenger) and chloride (saline) diuresis.

Platinum based, alkylating agents
• Mechanism of Action
– Forms intrastrand & interstrand DNA cross links
– Binds to nuclear and cellular proteins

• Toxicity
– Acute: Nausea & Vomiting (**highly emetogenic)
– Delayed: **Nephrotoxicity, **Ototoxicity, Peripheral sensory, & neuropathy
– **Irreversible nerve dysfunction

• Clinical Applications
– Non-small cell & Small cell lung cancers (NSCLC & SCLC)
– Head & Neck cancer
– Breast cancer
– Ovarian cancer
– Bladder cancer
– GE junction cancer
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Oxaliplatin

A

Platinum based alkylating agent
• Mechanism of Action
– Same as cisplatin and carboplatin but is effective in cells with DNA
Mismatch Repair (MMR) defects

• Toxicity
– Acute:
• Nausea and; Vomiting
• **Neurotoxicity — Laryngo-pharyngeal dysesthesia (Peripheral
sensory neuropathy which can be triggered by cold)
– Delayed: *Neurotoxicity—Peripheral sensory neuropathy which is dose-
dependent (
reversible unlike cisplatin), Diarrhea

• Clinical Applications
– **Colorectal Cancer (Component of “FOLFOX” regimen)
– Gastro-esophageal cancer
– Pancreatic cancer

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

Methotrexate

A
Folic acid analog
– Binds to and inhibits:
Reduced folate carrier
• DHFR (dihydrofolate
reductase) 
•  TS (thymidylate
synthase)

• Metabolized in the cell
– Polyglutamate metabolites
– Folylpolyglutamate synthase
-polyglutamates are selectively retained within cancer cells

• Disrupts the folate
metabolic pathway

Pharmacology
• Availability: IV or intrathecal (oral is poorly bioavailable)
• Biologic effects reversed by administration of the reduced folate–**LEUCOVORIN
• Renal excretion
– Dose adjustment with renal insufficiency
– ASA, NSAIDs, & penicillin inhibit renal excretion

• Toxicity
– Mucositis, diarrhea
– Myelosuppression—neutropenia & thrombocytopenia

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

Four mechanisms of MTX resistance

A

①Decreased drug transport

②Decreased formation of cytotoxic MTX polyglutamates

③Increased levels of the target enzyme DHFR

④Altered DHFR protein with reduced MTX affinity

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

5-fluorouracil (5-FU)

A
Parent form inactive 
• Activated metabolites include FdUMP (DNAi) and FUTP (RNAi) 
• FdUMP* inhibits TS
– Inhibits DNA synthesis by inducing
“thymineless” death

• FdUTP**
– Inhibits DNA synthesis and function

• FUTP***
– Interferes with RNA processing and
translation

Metabolism
– Administered IV; not orally available
– DPD is in saliva
– Short t1/2
– 85% of dose is catabolized by dihydropyridine dehydrogenase (DPD)
– Pharmacogenetics (5%) **If DPD is low or absent (DPD deficiency), toxicity is severe or life-threatening!

• Toxicity
– Myelosuppression
– (GI)-nausea, vomiting, diarrhea, mucositis
– Neurotoxicity
*Derm—“Hand-foot syndrome” with continuous infusion

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

Capecitabine

A
Weber designed this 
• PRO-PRO-DRUG of 5-FU 
• **Orally available 
• Active metabolites are generated
in the liver 
• Metabolites are converted to 5-
FU in the tumor
– Thymidine phosphorylase 

• Toxicity profile reflects continuous
infusion 5-FU particularly:
– *“Hand-foot syndrome” (Fluoro based medicines)
– Diarrhea
– {Not notorious for alopecia - other agents for breast cancer lead to alopecia}

• Clinical use in breast & Colorectal cancer

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

Cytarabine (Ara-C)

A
• **S-phase specific
antimetabolite 
• Competitively inhibits DNA polymerases
– Blocks DNA synthesis &
repair

• Incorporated into DNA
– Blocks chain elongation &
DNA ligation

IV Administration
– Cleared very rapidly; requires almost constant infusion
• Activity
– Hematologic malignancies exclusively
– AML- first-line [combination with idarubicin]
– Liposomal version indicated for lymphomatous meningitis

• Toxicity
– Acute: (GI)-Mucositis, nausea, vomiting, diarrhea
– Chronic: (**Heme)-Myelosuppresion (pancytopenia)

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

Gemcitabine

A

①Inhibits DNA polymerases
②Blocks DNA (& RNA) synthesis and repair (ribonucleatide reductase)
③Once incorporated, causes chain
(strand) termination

Activity
– Used against **broad range of malignancies:
– NSCLC 
– Pancreatic cancer 
– Bladder Ca 
– Ovarian Ca 
– NHL
• Toxicity
– Heme—**Neutropenia
– (GI)—Nausea & vomiting in 70% of patients
– “flu-like” syndrome
– Rare: ***Renal microangiopathy
 **Hemolytic Uremic Syndrome (HUS)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

6-Mercaptopurine (TPMT, allopurinol)

A

• Inactive parent molecule • Metabolized by HGPRT*
(Hypoxanthine-guanine
phosphoribosyl transferase)

• Inhibits enzymes of Purine
nucleotide synthesis

• Triphosphate forms can be incorporated into RNA and DNA Inactive metabolite

Deficiency of TPMT can lead to increased toxicity

6-MP is deactivated in
reaction catalyzed by
xanthine oxidase
• Allopurinol is often used for
supportive care: – Potent XO inhibitor – Can lead to excessive toxicity
when used with 6-MP – Requires dose reduction

• Pharmacogenetics
– Thiopurine methyl transferase (TPMT) deficiency can result in severe toxicities
• Myelosuppression
• Diarrhea

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

Vinblastine, vincristine, Vinorelbine

A
Vinca alkaloids that bind
β-tubulin and inhibit
its polymerization into
microtubules >  prevent
mitotic spindle formation
(M-phase arrest).

Uses: Solid tumors, leukemias,
Hodgkin (vinblastine) and
non-Hodgkin (vincristine)
lymphomas.

Vincristine: neurotoxicity
(areflexia, peripheral neuritis), constipation (including paralytic ileus).

Microtubule dysfunction (M phase) 
Vinca alkaloids: 
• Plant-based Alkaloid derivatives
• Inhibit microtubule
polymerization by binding to
b-tubulin to inhibit assembly
• Results in mitotic arrest in
metaphase

Vinblastine
• Metabolism
Liver cytochrome p450 system

• Toxicity
– Nausea, vomiting
– Bone marrow suppression
– Alopecia
– *POTENT VESICANT (blistering
agent)

Vincristine
• Active vs. hematologic
malignancies

• Toxicity
– ***Neurotoxicity with peripheral
neuropathy 
– **Autonomic system dysfunction
• Orthostatic hypotension 
• Urinary retention 
• Paralytic ileus
– Myelosuppression
– ***SIADH
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Taxanes

– Paclitaxel + protein bound

A

Mechanism: Hyperstabilize polymerized microtubules in M phase so that mitotic spindle cannot break down (anaphase cannot occur).

Clinical use: Ovarian and breast carcinomas.

Adverse effects: Myelosuppression, neuropathy,
hypersensitivity.
----------------------------------
• Microtubule spindle
poison that enhances
microtubule polymerization 

• Metabolism: cytochrome
p450 system so that dose
adjustment is necessary
for liver dysfunction

• Clinical use: broad range of
*solid tumors esp. breast
and ovarian cancers &
NSCLC

Paclitaxel: Toxicity
• Acute
– Nausea, vomiting, hypotension
– ***HYPERSENSITIVITY (5%) (in Cremophor based formulations)- can
be prevented with premedication with dexamethasone and diphendydramine

• Delayed
– Myelosuppression
– **(Neurotoxicity), peripheral sensory neuropathy
– ***(Initially single sourced from bark of Pacific Yew tree)

(paclitaxel protein bound or albumin bound)
• Paclitaxel formulated with near nanoparticle size
albumin carrier

• Toxicity
– No paclitaxel related hypersensitivity
– Decreased toxicity overall (vs. paclitaxel)

  • FDA approved in:
  • Breast Ca
  • Non-small Lung Ca
  • Pancreatic Ca
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Etoposide

A

Etoposide
MECHANISM Etoposide inhibits topoisomerase II > increase DNA degradation.
CLINICAL USE: Solid tumors (particularly testicular and small cell lung cancer), leukemias, lymphomas.

ADVERSE EFFECTS: Myelosuppression, alopecia.

  • 50% available orally
  • Metabolized in the liver and also excreted renally
• Toxicities:
– Nausea, vomiting
– Hypotension
– Myelosuppression
– Alopecia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Camptothecins (Topotecan, irinotecan)

A

Irinotecan, topotecan
MECHANISM Inhibit topoisomerase I and prevent DNA unwinding and replication.
CLINICAL USE Colon cancer (irinotecan); ovarian and small cell lung cancers (topotecan).
ADVERSE EFFECTS Severe myelosuppression, diarrhea.
Results in S phase arrest

Topotecan
• IV infusion
• Renal excretion

• Toxicity
– Nausea, vomiting
– Alopecia
– Bone marrow suppression
especially neutropenia

– Approved in Small Cell Lung Cancer

Irinotecan • PRODRUG converted in the liver
to SN-38 metabolite
– Metabolite is 1000x more potent
than irinotecan
• Excreted in the bile and feces 
• Toxicity
– ***********DIARRHEA into a teacan
• Early- cholinergic event • Late- 2-10d post treatment, can be
severe!
– Bone marrow suppression 

– First-line drug for CRC (FOLFIRI regimens)

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

Anthracycline (doxorubicin)

A

Generate free radicals.
Intercalate in DNA > breaks in DNA > decrease replication.

USE: Solid tumors, leukemias, lymphomas.

ADVERSE EFFECTS: Cardiotoxicity (dilated
cardiomyopathy),
myelosuppression, alopecia.
*Dexrazoxane (iron chelating
agent), used to prevent
cardiotoxicity.

Isolated from a soil microbe:
Streptomyces peucetius
Widely used cytotoxic drugs

example doxorubicin (big red)

①Inhibit Topoisomerase II
②High affinity DNA intercalation
– Blocks both DNA & RNA synthesis

③Generate ***semi-quinone free-radicals
– Major mechanism of **cardiotoxicity

④Bind to cell membranes
– Alters fluidity & ion transport

Pharmacology
• IV Administration
• Metabolism: via Liver to active metabolite
• Toxicity
– (Heme)—Myelosuppresion with neutropenia
– (GI)—Mucositis
– **Cardiotoxicity
• Acute (2-3d): arrhythmias and conduction abnormality
• Chronic: **dose-dependent dilated cardiomyopathy - a specific ceiling. (BIG RED)
(>550mg/m2 cumulative—increased risk)
• Iron-chelator Rx can reduce events in high risk patients
– **Dexrazoxane

One of the most important
anti-cancer drugs • Wide Efficacy • Used in combination
therapy (e.g. ABVD*) for
Hodgkins lymphoma

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

Bleomycin

A
Bleomycin
Induces free radical formation
>  breaks in DNA strands.
USES: Testicular cancer, Hodgkin lymphoma.
Pulmonary fibrosis, skin
hyperpigmentation. Minimal
myelosuppression.

• Glycopeptide antibiotic from Streptomyces verticillus
• Metal binding
• Binds to DNA
– Causes SS and DS breaks – Inhibits DNA synthesis – Lipid peroxidation

• Cell-Cycle specific: **G2 phase arrest

Metabolism
• IV, IM, or SQ infusion
• Renal excretion

• Toxicity
– PULMONARY

Uses: **Testicular cancer
Hodgkin’s, non-Hodgkin’s Lymphoma

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

Cell cycle specific to the drug classes

A

Cell cycle specific

Antimetabolite S

Epipodophyllotoxin G2-S

Anti-tumor Antibiotics
G1/S; phase G2

Taxanes M

Vinca Alkaloids M

Anti-microtubule inhibitor M

Bleomycin G2-M

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

Critzotinib

A

Crizotinib: (Inhibits ALK (activin receptor-like kinase), ROS1, HGFR and other TKs
• ~4% of NSCLC patients have translocations involving EML4- ALK1 resulting in constitutive kinase activity • Tend to be younger, non-smokers, wild type for EGFR
and RAS
• Indications: –ALK 1 (with companion diagnostic test for ALK) –ROS 1 mutations

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

Thalidomide, Lenalidomide, pomalidomide

A

Thalidomide

Originally developed as an anti-emetic/sedative: found to be teratogenic
(Phocomelia)
Revived when it was found to have potent anti-inflammatory effects in treatment of erythema nodosum leprosum

thalidomide, lenalidomide, pomalidomide)
Marked activity in Multiple Myeloma

22
Q

Bortezomib

A

Proteosome Inhibitor: Bortezomib

Proteosome inhibitors initially developed for treatment of cachexia

Proteosome-mediated degradation was found to be a key regulator of multiple signaling pathways.

Indication: Multiple Myeloma

Toxicities: Neuropathy, myelosuppression

23
Q

Bevacizumab

A

MECHANISM Monoclonal antibody against VEGF. Inhibits angiogenesis (BeVacizumab inhibits Blood Vessel formation).
CLINICAL USE Solid tumors (colorectal cancer, renal cell carcinoma).
ADVERSE EFFECTS Hemorrhage, blood clots, and impaired wound healing.

Bevacizumab

Limited single agent activity in solid tumors (lung, colon, breast).
Improved clinical outcome when added to standard chemotherapy in treatment of advanced lung, colon and breast cancer. Improved overall survival in lung cancer, but risk of pulmonary hemorrhage high in patients with squamous cell cancers.
Also active in ovarian and renal cell cancer.
Highly active in neo-vascular diseases of the retina.

Toxicities: Hypertension, pulmonary hemorrhage, proteinuria

24
Q

PD1/PDL-1 checkpoint inhibitors (nivolumab, pembrolizumab, atezolizumab)

A

PD-1 inhibitors blocks the interaction of the tumor and the T-cell

1) Binds to programmed death receptor-1 (PD-1) which inhibits programmed T cell death activity
2) Blocks the PD-1 pathway to inhibit the negative immune regulation caused by PD-1 receptor signaling
3) Reverses T cell suppression and induces antitumor response

• Nivolumab- PD-1 Inhibitor
– Hodgkin lymphoma, melanoma, NSCLC, renal cell carcinoma, head and neck cancer, urothelial carcinoma

• Pembrolizumab- PD-1 Inhibitor
– Melanoma, **NSCLC (only first line), head and neck cancer, urothelial carcinoma

• Atezolizumab- PDL-1 Inhibitor
– Urothelial carcinoma

25
Q

Alemtuzumab

A

Alemtuzumab

Humanized monoclonal antibody that binds to CD52 present on surface of mature lymphocytes (both B and T cells) undergoing cellular lysis

Approved for treatment of B-cell CLL (and is used for the
treatment of a variety of T-cell malignancies).

Side effects: prolonged T-cell depeletion/immunosupression

Black box warning: infusion reactions, cytopenia

26
Q

Imatinib (FA)

A

FA
MECHANISM Tyrosine kinase inhibitor of BCR-ABL (Philadelphia chromosome fusion gene in CML) and c-kit
(common in GI stromal tumors).
CLINICAL USE CML, GI stromal tumors (GIST).
ADVERSE EFFECTS Fluid retention.

TK inhibitor
• Binds to catalytic cleft of ABL
• Remarkably effective in treating CML as a single agent
• Leads to high rate of long-term cytogenetic remission

Indications:
1) Chronic Myelogenous Leukemia (CML):
–Characterized by 9:22 translocation
–Translocation creates BCR-ABL fusion protein—a constitutively active
TK
2) GIST: c-KIT is overexpressed/mutant in GIST
(gastrointestinal stromal tumor)–systemic mastocytosis

Major Toxicities
Edema (face, hands, and feet)
Skin rash

27
Q

Trastuzumab (FA)

A

MECHANISM Monoclonal antibody against HER-2 (c-erbB2), a tyrosine kinase receptor. Helps kill cancer cells that overexpress HER-2, through inhibition of HER2-initiated cellular signaling and antibodydependent
cytotoxicity.
CLINICAL USE HER-2 ⊕ breast cancer and gastric cancer (tras2zumab).
ADVERSE EFFECTS Cardiotoxicity. “Heartceptin” damages the heart.

A humanized (not human) IgG 1 kappa monoclonal antibody
Mechanism of action:
1. Interferes with HER2-dependent signaling
2. Antibody-dependent cellular cytotoxicity (immune mediated)
Indication: HER2+ breast cancer
(Induces response in up to 50% of HER2+ breast cancers)
Major side effect: Cardiac toxicity

28
Q

Erlotinib (FA)

A

MECHANISM EGFR tyrosine kinase inhibitor.
CLINICAL USE Non-small cell lung carcinoma.
ADVERSE EFFECTS Rash.
————————————–
• Small molecule TKI

• Responses in 10% of NSCLC; all responders had EGF-R mutations
{Most commonly responders are women, non-smokers,
broncheoalveolar histology}

Major Toxicities

  • **Derm: Rash (but indicates medication is working)
  • GI: Nausea, Vomiting, Diarrhea
  • Pulmonary: Cough & Shortness of breath
29
Q

Cetuximab (FA)

A

Cetuximab
MECHANISM Monoclonal antibody against EGFR.
CLINICAL USE Stage IV colorectal cancer (wild-type KRAS), head and neck cancer.
ADVERSE EFFECTS Rash, elevated LFTs, diarrhea.
————————————

-Clinical activity:
• Advanced colorectal cancer
• Advanced Head and neck cancer
(K-RAS mutations and BRAF mutations are associated with lack of response to Cetuximab)
-Radiation sensitizer
30
Q

Vemurafenib (FA)

A

MECHANISM Small molecule inhibitor of BRAF oncogene ⊕ melanoma. VEmuRAF-enib is for V600Emutated
BRAF inhibition.

CLINICAL USE Metastatic melanoma.

-Small molecule RAF inhibitor
-**Targets V600E BRAF mutation
Cancers with B-RAF mutations:
Melanoma, NHL, colon cancer, papillary thyroid
cancer, non-small cell lung cancer, hairy cell leukemia
Major Toxicity:
Causes other skin cancers (cutaneous squamous
cell and keratoacanthomas)

Addition of MEK inhibitor (tremetinib) prevents other skin cancers induced by
BRAF inhibitor

31
Q

Tamoxifen, raloxifene (FA)

A

—used in breast cancer especially for
multi-year prophylaxis

MECHANISM Selective estrogen receptor modulators (SERMs)—receptor antagonists in breast and agonists in
bone. Block the binding of estrogen to ER ⊕ cells.

CLINICAL USE Breast cancer treatment (tamoxifen only) and prevention. Raloxifene also useful to prevent
osteoporosis.

ADVERSE EFFECTS Tamoxifen—partial agonist in endometrium, which increases the risk of endometrial cancer; “hot flashes.”
Raloxifene—no increases in endometrial carcinoma (so you can relax!), because it is an estrogen receptor
antagonist in endometrial tissue.
Both increases risk of thromboembolic events (eg, DVT, PE).

32
Q

Rituximab (FA)

A

MECHANISM Monoclonal antibody against CD20, which is found on most B-cell neoplasms.
CLINICAL USE: Non-Hodgkin lymphoma, CLL, ITP, rheumatoid arthritis (autoimmune).
ADVERSE EFFECTS Increases risk of progressive multifocal leukoencephalopathy.
——————————————–
Chimeric monoclonal antibody against CD20
Transmembrane protein found on the surface of pre-B cells and mature B cells

Highly effective in treatment of some B-cell malignancies, and is now part of standard treatment of B-cell NonHodgekinLymphoma
{Also now widely used
in autoimmune disease}

Toxicities: multifocal leukoencephalopathy
flushing, fever, chills,
nausea, diarrhea

33
Q

Tumor lysis syndrome

A

Oncologic emergency triggered by massive tumor cell lysis, most often in lymphomas/leukemias.

Release of K+ > hyperkalemia, 
Release of PO4
3– > hyperphosphatemia, 
Hypocalcemia due to
Ca2+ sequestration by PO4
3–. INcreased nucleic acid breakdown > hyperuricemia > acute kidney injury.

Treatments include aggressive hydration, allopurinol, rasburicase.

34
Q

Common chemotoxicities

of 
Cisplatin/Carboplatin
Vincristine
Bleomycin, Bulsulfan
Doxorubicin
Trastuzumab
Cyclophosphamide
A
Cisplatin/Carboplatin >  ototoxicity (and
nephrotoxicity)
Vincristine > peripheral neuropathy
Bleomycin, Busulfan > pulmonary fibrosis
Doxorubicin > cardiotoxicity
Trastuzumab (Herceptin) > cardiotoxicity
Cisplatin/Carboplatin > nephrotoxicity (and
ototoxicity)
CYclophosphamide > hemorrhagic cystitis
35
Q

Hydroxyurea

A

MECHANISM Inhibits ribonucleotide reductase > decreased DNA Synthesis (S-phase specific).
CLINICAL USE Myeloproliferative disorders (eg, CML, polycythemia vera), sickle cell (increased HbF), melanoma.
ADVERSE EFFECTS Severe myelosuppression.

36
Q

Oncodrugs that cause neurotoxicity

A
  1. Vincristine (microtubule inhibitor)
  2. Paclitaxel/Docetaxel (microtuble stabilizer)
  3. Cisplatin - peripheral sensory and neuropathy (irreversible)
  4. Oxaliplatin - laryngo-pharyngeal dysesthesia, and reversible peripheral sensory neuropathy
  5. 5-Fluorouracil - pyramidine inhibitor
  6. Bortezomib- proteosome inhibitor (used for multiple myeloma)
37
Q

Which onco drug has a toxicity of cardiotoxicity

A
  1. Doxorubicin - dose-dependent dilated cardiomyopathy (use iron chelator) - generates free radicals
  2. Trastuzumab - IgG 1 kappa monoclonal antibody - HER2 breast cancer
38
Q

Which onco drugs have pulmonary toxicities

A
  1. Bleomycin- binds to DNA, G2 phase arrest

2. Bevacizumab- risk of pulmonary hemorrhage

39
Q

Which onco drug has a metabolite that acts as a monoamine oxidase inhibitor

A
  1. Procarbazine
40
Q

Which onco drugs are used for breast and ovarian cancers

A
  1. cyclophoshamide - nitrogen mustard alkylating agent
  2. Taxanes: breast and ovarian
  3. Trastuzumab - HER2 + breast cancer
  4. Tamoxifen - used in breast cancer multiyear prophylaxis
41
Q

Which onco drugs are used mainly for NSC or SC lung cancers?

A
  1. cisplatin
  2. Taxanes - NSCLC
  3. Erlotinib, TK inhibitor (NSCLC)
  4. Crizotinib - NSCLC
  5. Nivolumab (PD-1 inhibitors)
42
Q

Which oncodrugs are used for Colorectal cancer

A
  1. Oxaliplatin - MMR defects best for

2. Cetuximab - EGF-R monoclonal antibody

43
Q

Oncodrugs that cause nephrotoxicity

A
  1. Cisplatin
  2. Oxaliplatin
  3. ATRA (renal failure)
44
Q

Which drug is first line for AML

A

Ara-C (Cytarabine) - inhibitor of DNA polymerase

45
Q

Which onco drugs cause diarrhea

A
  1. Irinotecan - very severe diarrhea (topo I inhibitor)
  2. 5-FU - pyrimidine inhibitor
  3. Capecitabine - proprodrug of 5-FU
  4. 6-mercaptopurine - purine synthesis inhibitor
  5. Erlotinib
46
Q

Which oncodrugs are known to cause edema?

A
  1. ATRA (capillary leak syndrome)

2. Imatinib (face hands and feet edema)

47
Q

Which onco drugs cause derm manifestations

A
  1. 5-FU (hand foot syndrome)
  2. Capecitabine (hand foot syndrome )
  3. Erlotinib, - tyrosine kinase inhibitor
  4. Vemurafenib - cuases other skin cancers, targets V600E BRAF mutation
48
Q

Which drugs treat melanoma

A
  1. Nivolumab, (PD-1 inhibitors)
  2. Ipilumamb- CTLA-4 inhibitor (severe immune reactions)
  3. Vemurafenib - BRAF inhibitor
49
Q

What is an oncodrug widely used to treat autoimmune disease

A

Rituximab - monoclonal antibody against CD20

77
Q

Rasburicase

A

rasburicase. This is because when the patient has TLS and uric acid levels are already HIGH- ALLOPURINOL will not be as effective in managing the acute elevation in uric acid.
Therefore, treatment with an agent that will LOWER URIC ACID levels is the best choice and rasburicase will do this by converting uric acid to allantoin.