• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/72

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

72 Cards in this Set

  • Front
  • Back
Cell cycle
G1 Growth occurs as organelles double
S DNA replication occurs as chromosomes duplicate
G2 Growth occurs as cell prepares to divide
M Mitosis and cytokinesis occurs
Cell cycle checkpoints
Cyclin D/cdk 4, 5, 6
-M phase
-when EGFR is upregulated, cycling D is upregulated
Classical chemotherapy: cell phase specific vs non cell phase specific
NON-Cell phase SPECIFIC
-Kill both normal and malignant cells in all phases of cell cycle
-Cell kill proportional to dose
-Bolus dosing
-Alkylating agents, anthracyclines

Cell Phase SPECIFIC
-Cytotoxic to part of cell cycle in which it is active
-Administer via continuous infusion
-Mitosis: vinca alkaloids, taxanes
-G1: steroids, asparaginase
-G2: bleomycin, etoposide, tenoposide
-S: antimetabolites, camptothecins
Sites of action of cytotoxic agents
Antimetabolites: DNA synthesis
Alkylating agents: DNA
Intercalating agents: DNA transcription, DNA duplication
Spindle poisons and microtubule stabilizers: Mitosis
Alkylating agents mechanism of action
One 2-chloroethyl side chain undergoes a 1st order intramolecular cyclization
Result is the release of Cl- and formation of a highly reactive aziridinium ion
Binds to DNA resulting in crosslinking or intrastrand
-usually on guanine
Alkylating agents classes
Nitrogen mustards
Ethylenimines
Alkly sulfonate
Nitrosoureas
Triazenes
Platinum analogues
Potential outcomes of alkylating agents
Template being replicated is misread or mismatched during DNA synthesis

Cross-linking prevents DNA strands from unwinding

Single or double-strand breaks in DNA occur

DNA molecule ineffective
Cyclophosphamide/Ifosfamide clinical uses
Nitrogen mustards
Cyclophosphamide
-Leukemia, lymphomas, solid tumors
-Bone marrow transplant (high doses > 2 g/m2)
-Treat graft-versus-host-disease
-Immunosuppression in nonmalignant diseases:
--Rheumatic disorders, lupus, & autoimmune nephritis

Ifosfamide
-Solid tumors: breast, colon, lung
-Testicular cancer
-Soft tissue sarcomas
Cyclophosphamide/Ifosfamide metabolism
Given in inactive form
Activated by hepatic metabolism by cytochrome p450
Either inactivated by aldehyde dehydrogenase or converted to acrolein or phosphoramide mustard
-acrolein: toxic, ifosfamide causes more acrolein buildup than cyclophosphamide
-phosphoramide mustard: cytotoxic, active agent
Hemorrhagic cystitis
Caused by accumulation of ACROLEIN
-Binds to thiol in bladder wall
Hematuria, urinary frequency & irritation
Prevent with vigorous hydration (≥2 L/day) & MESNA
Treat with bladder irrigation, alum irrigation, and other therapies
Heme test urine while on therapy
MESNA
Mercaptoethane sodium
Uroprotectant containing sulfhydryl group
-Binds to acrolein in the bladder to form a nontoxic compound
Not systemically absorbed so does not interfere with cytotoxic activity
MESNA is indicated in:
-Cyclophosphamide >1 g/m2/dose
-Ifosfamide at any dose
Effective in PREVENTION only
Cyclophosphamide/ifosfamide toxicities
Cyclophosphamide
-Hemorrhagic cystitis
-Delayed Nausea & vomiting
-Syndrome of inappropriate anti-diuretic hormone (SIADH)
-Pulmonary

Ifosfamide
-Hemorrhagic cystitis
-Neurotoxicity
-Renal Fanconi syndrome
Platinum agents
Alkylating

Cisplatin
-Renally cleared
-Nephrotoxicity – ↓ GFR, electrolyte losses (Mg, K), and renal failure
--Prevent with aggressive hydration (NaCl)

Carboplatin
-Not concentrated in the renal tubules; more efficiently cleared
-Dosing based on area under the curve (AUC)
--Calvert Equation: Dose = AUC ( GFR + 25 )
(GFR=renal function)

Oxaliplatin
Platinum analogues uses
Breast
Lung
Testicular
Gyn/Onc: cervical and ovarian
Colorectal
Bladder
Lymphoma
Platinum analogues toxicities
Cisplatin
-**Acute/Delayed Nausea/Vomiting
-**Nephrotoxicity
-Peripheral neuropathy
-Neurotoxicity
-Ototoxicity

Carboplatin
-**Myelosuppression - ↓ platelets
-Neurotoxicity
-Vomiting

Oxaliplatin
-**Peripheral neuropathy
-Myelosuppression
Uncommon alkylating agents
Nitrosoureas
-Carmustine*
-Lomustine
-Streptozocin
-Very liphophilic
-Brain tumors

Triazenes
-Dacarbazine
-Temozolomide*
-Melanoma, lymphoma, glioma, and sarcoma
Common oral alkylating agents
Chlorambucil (Leukeran)
-Used for the treatment of chronic lymphocytic leukemia

Busulfan (Myleran)
-Used for treatment of leukemia and transplant
-Also available in an IV form (used more for transplant)
-Toxicity is decreasing seizure threshold

Melphalan (Alkeran)
-Used for the treatment of multiple myeloma
-8 mg/m2 PO daily day 1-4 repeat every 28 days

All three of these medications come as 2 mg tablets (watch dispensing by brand name, avoid wrong drug)
Alkylating agents: common toxicities
Myelosuppression
-Prolonged/delayed myelosuppression nadir
Secondary leukemias
Infertility
Alopecia
Nausea/vomiting
Topoisomerase inhibitors
Topoisomerase I inhibitors
-Camptothecins
--Irinotecan
--Topotecan*
Topoisomerase II inhibitors
-Anthracyclines
--Daunorubicin
--Doxorubicin
--Idarubicin
--Epirubicin
-Epipodophyllotoxins
--Etoposide*
--Teniposide*
-Miscellaneous
--Mitoxantrone
Topoisomerase inhibitors mechanism of actions
Topo I: 1 nick in DNA
Topo II: 2 nicks in DNA

Won't let nicks be resealed
Anthracyclines
Topoisomerase II inhibitors
-Cuts both strands of one DNA double helix can't be resealed

Things that end in -rubicin
-Daunorubicin
-Doxorubicin
-Idarubicin
-Epirubicin
-Miscellanous:
--Mitoxantrone
Anthracyclines clinical uses
Doxorubicin/Epirubicin
-Breast cancer
--Most active agent
-Sarcomas
-GI tumors
-Lymphoma

Daunorubicin/Idarubicin
-Leukemia

Mitoxantrone
-Leukemia
-Prostate cancer
Anthracyclines toxicities
**Myelosuppression
**Cardiotoxicity
**Extravasation injury
-Vesicant
-Treat with dexrazoxane (Totect) and cold compress
Nausea and vomiting
Mucositis
Red/orange urine discoloration
Radiation recall
Anthracyclines cardiotoxicity mechanism of action
Increased oxygen derived free radicals
Damage lipid proteins and DNA predominantly seen in heard
Anthracyclines: cardiotoxicity cumulative dosing
Doxorubicin cardiotoxicity threshold
-Von Hoff – 7% in patients who receive 550 mg/m2
-Swain data – 26% in patients who receive 550 mg/m2
Patients who receive doses > 350 mg/m2should be treated with caution
Anthracyclines: cardiotoxicity classificaiton
Acute
-Immediate – after a single dose or course of therapy
-Decreased contractility, Pericarditis, Myocarditis,
Arrhythmia, Elevation of biomarkers

Early onset progressive
-Within 1 year of therapy with rapid onset and progression
-Tachycardia, ventricular dilation, exercise intolerance, pulmonary and venous congestion

Late-onset chronic progressive
-after 1 year of therapy completion
-ventricular dysfunction, conduction disturbances, arrhythmias, CHF
Cardiotoxicity prevention
Dexrazoxane
-Inhibit the iron-dependent free radical formation that can cause myocardial damage

Liposomal doxorubicin (Doxil®)
-Liposomal delivery system not as readily taken up by cardiac tissue: decreased risk of cardiotoxicity
-Used in breast, ovarian cancer
Mitoxantrone
Related to anthracyclines

Does not form free radicals

Lifetime dose = 180mg/m2

↓ cardiotoxicity and extravasation

↓ nausea and vomiting

Blue-green urine discoloration
Epipodophyllotoxins
Etoposide (VP-16) & Teniposide (VM-26)
Inhibit topoisomerase II
Toxicities
-**Myelosuppression
-Mucositis (BMT)
-Hypotension (diluent)

Clinical uses
-Etoposide – leukemias, NHL, BMT, small cell lung cancer (SCLC) , ovarian
-Teniposide* – ALL, SCLC
Camptothecins
Irinotecan & topotecan

Inhibit topoisomerase I

Clinical uses
-Ovarian cancer
-Lung cancer
-Cervical and ovarian cancer
-Colorectal cancer
Camptothecins toxicities
Irinotecan:
Severe diarrhea (20%)
-Acute (≤ 24 hours)
-Atropine
Chronic (~11 days)
-Loperamide

Topotecan:
Myelosuppression
-Leukopenia
-Thrombocytopenia
Methotrexate clinical uses
Clinical uses:
Osteosarcoma
Acute Lymphocytic Leukemia
Non-Hodgkins and CNS Lymphomas
Breast/bladder cancer
Non-oncologic uses
Methotrexate mechanism of action
Antimetabolite
Reduced folate analog

Taken up intracellularly by cancer & healthy cells

Inhibits DHFR: decreases tetrahydrofolate (THF): decrease purine & thymidylate

Lack of purines & thymidylate prevents DNA synthesis
Leucovorin (folinic acid)
Required for high dose MTX is lethal unless rescue initiated

Directly converted into tetrahydrofolate
-Does not require DHFR

Allows resumption of DNA synthesis even in the presence of MTX
Methotrexate toxicities and risk factors
Nephrotoxicity
Myelosuppression
GI toxicity
Hepatotoxicity
Neurotoxicity
Dermatitis
Death

Risk factors:
Pre-existing renal dysfunction
Urine pH < 7
Concomitant medications
Down syndrome
Third space fluids
-Ascites
-Pleural effusions
Methotrexate drug interactions
Sulfonamide: compete with MTX for transport and excretion
PCN: alters urine pH
NSAID: decrease renal blood flow, decrease excretion
PPI: inhibit H-K ATPase pump in renal tubules
Prevention of methotrexate toxicities
Renal function (CrCl > 60ml/min)

Vigorous hydration and alkalinization of the urine
-Administer 3,000 mL/m2/day (125mL/m2/hour) + 30-60 mEq/L
-pH > 7 = MTX solubility is pH dependent
-Urine specific gravity <=1.01

Avoid interacting medications

Leucovorin rescue with high doses (>1g/m^2)

Glucarpidase will cleave leucovorin and stop it from working
Pemetrexed (alitma)/Pralatrexate (Folotyn)
Antimetabolites

Pemetrexed:
-Multitargeted antifolate
-Clinical use:
--Malignant pleural mesothelioma
--NSCLC
-Toxicities:
--Cutaneous reaction
--FA/B12 supplement required

Pralatrexate:
-Folic acid analogue
-Clinical use:
--Relapsed/refractory peripheral T-cell lymphoma
-Toxicities:
--Mucositis
- -platelets/WBC
--FA/B12 supplement required
Cytarabine overview and mechanism
Antimetabolite
Arabinose analog of cytosine
Phosphorylated to active
component within cancer cells
Inhibits DNA polymerase
Cytarabine clinical uses
Acute leukemias
-Acute myeloid leukemia
-Acute lymphoblastic leukemia
-Meningeal leukemia

No significant activity against solid tumors
Cytarabine toxicities
Induction state:
(100 mg/M2/day)
-**Myelosuppression
-Alopecia
-Gastrointestinal
-Rash—palmer plantar syndrome

Consolidation:
(1-3 g/M2 q12h)
-**Nausea
-**CNS toxicity
-**Chemical conjunctivitis
-**Rash
Gemcitabine (Gemzar)
MOA & structure similar to cytarabine

Effective for solid tumors
-Pancreatic cancer
-Non-small cell lung cancer
-Sarcomas
-Bladder cancer

Intracellular concentrations 20x > cytarabine
Gemcitabine (Gemzar) toxicities
Myelosuppression
Generalized rashes
Fever and flu-like symptoms
Peripheral edema
Nausea and vomiting-mild
NOT Neurotoxic
Clofarabine and Nelarabine
Pyrimidine antagonists
Clofarabine (Clolar)
-Relapsed pediatric ALL or relapsed adult AML
-AE-skin toxicity-rash to desquamation

Nelarabine (Arranon)
-T-cell ALL or T-cell lymphoblastic lymphoma
-AE-neurotoxicity
Fluorouracil: mechanism of aciton
Fluorinated analog of uracil
Metabolized to FdUMP
Inhibits thymidylate synthase

Leucovorin synergistically helps fluorouracil by locking in FdUMP and not letting it pop off
Fluorouracil: clinical use
Treatment of solid tumors including breast, colorectal and other GI tumors

Non-oncologic uses: actinic keratoses and noninvasive skin cancers
-Cream formulation
Capecitabine (Xeloda)
Oral prodrug of fluorouracil
Metabolized to active component in tumor tissue
Use in metastatic colorectal & breast cancer
5-FU and capecitabine: toxicities
**Myelosuppression (bolus)
**Bloody diarrhea and hand foot sydrome (CI)
**Mucositis (CI)
Dermatologic
Ocular
Nausea and vomiting (mild)
Cardiotoxicity (rare)
Purine analogs
Inhibit de novo purine synthesis
Clinical use: leukemias
Mercaptopurine (6-MP)
Thioguanine (6-TG)
Fludarabine & cladribine
-Immunosuppressive T-helpher cells
-Prophylactic meds required
Vinca alkaloids: MOA
Mitotic inhibitors
Plant products derived from periwinkle plant
Prevent the assembly of tubulin dimers into microtubules
Cells accumulate in mitosis
Vinblastine, vinorelbine, vincristine uses and toxicities
Vinca alkaloid, mitotic inhibitor

Vinblastine
Uses:
-Hodgkin’s disease Lymphomas
-Breast
-Testicular tumors
Toxicity:
-Bone marrow
suppression, anorexia,
nausea,/vomiting &
Diarrhea, Alopecia

Vinorelbine:
Uses :
-NSCLC
-Breast
-Lymphoma
-Ovarian
Toxicity:
-Bone marrow
suppression, anorexia,
Nausea/ vomiting &
Diarrhea, Alopecia

Vincristine:
Uses:
-Childhood leukemias
-Childhood tumors-Wilm’s tumor, Neuroblastoma, Hodgkin’s disease
Toxicity:
-Peripheral neuropathy with
Paresthesia, Muscle weakness
***Vincristine has marrow sparing effect

All agents are vesicants and fatal if given intrathecally
Taxanes: MOA
Mitotic inhibitor
Promote microtubule assembly
Interfere with microtubule disassembly
Taxanes: clinical uses
Ovarian cancer
Breast cancer
Melanoma
NSCLC
Prostate cancer
Head and Neck cancer
Taxanes: toxicities
**Myelosuppression
**Mucositis
Peripheral neuropathy (cumulative)
Alopecia
Hypersensitivity reactions*
Nausea and vomiting (rare)
*premedicate with dexamethasone, H1 and H2 antagonist

Paclitaxel
-Myalgia
-Bradycardia
-Cremaphor EL

Docetaxel
-Fluid retention
-Palmar-plantar rash
-Polysorbate-80

Cabazitaxel
-Myelosuppression
-Diarrhea
-Fatigue
-Ethanol/polysorbate 80
Ixabepilone (Ixempra) MOA
Mitotic inhibitor
Treatment of patients with metastatic or locally advanced breast cancer:
Binds multiple ß-tubulin isoforms, including ßIII-tubulin to inhibit microtubule dynamics
-Overexpression of ßIII is associated with in vivo and clinical resistance to taxanes
Ixabepilone: adverse events
Similar to taxane

Neurotoxicity

Neutropenia

Infusion related reactions
Erubulin (Halaven): MOA
Inhibits formation of mitotic spindles
-Unique mechanism, no cross-resistance with taxanes
Indicated for treatment of 3rd-line breast CA
Erubulin (Halaven) Adverse effects
Neutropenia

Alopecia

Fatigue

Peripheral neuropathy – secondary to previous treatments?
Asparaginase: MOA, use, toxicities
Degrades asparagine found in the serum

Used for leukemia and lymphoma

3 formulations

Toxicities:
-Allergic reactions
-Pancreatitis
-fibrinogen
Treatment for acute promyelocytic leukemia
APL
-t(15;17) fuses PML gene with retinoic acid receptor-a (RAR-a)

All trans-retinoic acid (ATRA) releases this repression and allows promyelocytes to differentiate

Arsenic trioxide (ATO)
-QTC prolongation – monitor electrolytes

Retinoic acid syndrome (both agents)
-Fever, dyspnea, pleural effusion, peripheral edema, hypotension
-Treatment: dexamethasone 10 mg IV BID x 3 days
Proteasome inhibitors MOA
Prevent activation of transcription by NFkappabeta
Bortezomib (Velcade): indications, Toxicities, administration
proteasome inhibitor

Indications:
-Multiple Myeloma
-Mantle cell lymphoma

Toxicities:
-Peripheral neuropathy
-Thrombocytopenia
-Neutropenia
-Fatigue
-Weakness
-GI effects

Intravenous versus subcutaneous administration
Carfilzomib (Kyprolis): indicaiton, toxicities
proteasome inhibitor

Indication: Relapsed/refractory multiple myeloma in patients who have failed therapy with bortezomib and an immunomodulatory agent

Toxicities
-Cardiac arrest/congestive heart failure
-Pulmonary hypertension/dyspnea
-Thrombocytopenia
Antibiotics (mycin)
Bleomycin
-Testicular cancer and Hodgkin’s Disease
-Watch for pulmonary toxicity and N/V

Dactinomycin
-Wilms tumor and rhabdomyosarcoma
-N/V, decreased counts

Mitomycin C
-Gastrointestinal tumors
-Intravesicularly in bladder cancer
Hydroxyurea (hydrea)
Inhibits ribonucleotide reductase
Used primarily for CML
Toxicity:
-Myelosuppression

Non-oncology indications: Sickle cell disease
Immunomodulators
Thalidomide – for multiple myeloma (MM)
- increased Thromboembolism
-Drowsiness/somnolence
-Peripheral neuropathies

Lenalidomide (Revlimid)-for MDS and MM
-Myelosuppression

Must have pregnancy test and contraceptive use
Histone deacetylators inhibitors MOA
Histone is a protein important for DNA packing

Acetylation = (HAT)
-Control tightness DNA binding
Deacetylation (HDAC)
-catalyzes remove acetyl groups
-Inhibit gene expression

Allows the cell to develop normally

Cancer cells  HDAC allows the cell to grow unregulated

HDAI cause accumulation of acetyl groups

Induce cell cycle arrest or apoptosis
Histone deacetylators inhibitors: clinical use, toxicity
Clinical Use
-Cutaneous T-cell lymphoma (CTCL)
--Cutaneous manifestations in patients
--Progressive, persistent or recurrent disease on or following two systemic therapies
-Peripheral T-cell lymphoma (PTCL) – romidepsin only

Vorinostat – PO only
Romidepsin – IV only

Toxicities:
-Thrombotic events/QTc prolongation
-Myelosuppression: decreased platelets/Hgb
-GI: nausea/vomiting/diarrhea
Hypomethylating agents
Incorporates into DNA and inhibits transfer of methyl groups
Hypomethylation = leads to cancer cell death

Azacitidine (Vidaza) & Decitabine (Dacogen)
-Indication: Myelodysplastic syndrome
-Decitabine also used in AML

Dose limiting toxicity: Myelosuppression
Hedgehog pathway inhibitor MOA
PTCH1 inhibits the release of SMOH receptor
Activation of HH pathway downregulates PTCH1
End result – promotes transcription and DNA replication
Vismodegib
Hedgehog pathway inhibitor

Binds to PTCH1 and blocks release of SMOH receptor to cell wall

Used in basal cell carcinomas

Adverse effects:
-Alopecia
-Muscle spasms
-Nausea/vomiting
-Embryofetal death
Corticosteroids, hormonal agents
Block hormones
Tumors promoted by amount of hormone in body