• 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

1/88

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;

88 Cards in this Set

  • Front
  • Back
The cells kill by releasing small cytoplasmic granules of proteins called?
Perforin and Granzyme
NK cells are activated by?
interferon-γ
Antibodies are recognized by ____ receptors on cells and phagocytes
Fc receptors
tumor-specific antigens (TSA) expressed by tumor cells but
not expressed by normal cells?
Bcr-Abl
Cancer caused by the Philadelphia Chromosome?
chronic myelogenous leukemia (CML).
CD8 cells must be activated by?
Th or CD4 cells
Tumor infiltrating lymphocytes (TILs) are mainly?
cytotoxic T cells (Tc, CTLs, CD8+)
Tumor cells display their own antigen on?
MHC-I
Regulatory T cells (T-regS)?
CD4+ and CD25+
Regulatory T cells inhibit both?
inhibit both Th (CD4+, CD25-) and Tc (CTL, CD8+ CD25-) effector cells
There is a strong correlation between the presence of _____ and the survival of patients with many cancers (e.g. ovarian, melanoma, breast,
colorectal)
TILs
TILs indicate a strong _______?
Cellular response
Tumor cells' primary mechanism of resistance?
Decrease the expression of MHC-I
Tumor cells secrete these cytokines to inhibit immunocytes?
TGF-β and IL-10
Four Immunity-Mediated Cancer Therapies?
- immunostimulants (IFN-α, IL-2)
- cancer vaccines (in clinical trials)
- adoptive T-cell transfer (in clinical trials)
- monoclonal antibodies
MOA of Interferon-alfa (IFN-α)?
stimulates cytotoxic activity of CTLs, NK cells and macrophages
• upregulates MHC class 1 and other surface markers
Major hurdle in adaptive T-cell transfer?
establishment of stable population of reintroduced TILs in vivo
What are the 5 classes of alkylating agents?
1. nitrogen mustards
2. ethyleneimines
3. alkyl sulfonates
4. nitrosoureas
5. triazenes
What are the 6 nitrogen mustard agents?
cyclophosphamide, ifosfamide, chlorambucil, mechlorethamine,
melphalan, bendamustine
What are the 2 most important nitrogen mustard agents?
cyclophosphamide and ifosfamide
Name the alkyle sulfonate alkylating agent?
busulfan
Name the triazene alkylating agents?
dacarbazine and procarbazine
Functional group of nitrogen mustard agents?
bis-(2-chloroethyl)
Mechanism of Alkylating Reaction of Nitrogen Mustard?
Binding to the N7 position of guanine, causing interstrand cross-linakge
Cyclophosphamide is used to treat?
lymphoma, breast cancer, acute lymphocytic leukemia (ALL) and ovarian cancer
Cyclophosphamide is a prodrug that makes the toxic metabolite _______?
Acrolen
Hemorrhagic cystitis caused by Cyclophosphamide can be counteracted by?
Mesna (sulfonate) and diuresis
Severe toxicity caused by Ifosfamide?
CNS toxicity (crosses the BBB)
Cell cycle effected by platinum complexes?
Platinum complexes are cell-cycle non-specific
Name the 3 platinum complexes?
cisplatin, carboplatin and oxaliplatin
Platinum complexes react with DNA forming intrastrand and interstrand cross-links, with N7 of guanine as the primary target. What is the favored cross-linking mechanism?
Intrastrand
Platinum complexes are cell-cycle non-specific, but they are most effective during ___phase
S-phase
Cisplatin causes nephrotoxicity that can be alleviated by?
chloride diuresis and amifastine (Ethyol)
pyrimidine analogs
5-FU, capecitabine, cytarabine, gemcitabine, azacytidine,
decitabine
purine analogs
6-mercaptopurine, fludarabine, pentostatin, cladribine,
clofarabine, nelarabine
Phase of cell cycle affected by antimetabolites?
All antimetabolites are S-phase specific
Primary MOA of MTX?
inhibition of DHFR
MTX toxicity can be rescued by?
Leucovarin (folinic acid, N5-formyl FH4) a reduced folate coenzyme
MOA of hydroxyurea?
Inhibition of ribonucleotide reductase, thus blocking DNA synthesis
MOA of 5-FU?
blocks synthesis of TMP by inhibiting THYMIDYLATE synthase
Agent that enhances the activity of 5-FU?
Leucovarin
MOA of purine analogs?
inhibition of de novo synthesis of purine bases and nucleotides and as triphosphate derivatives incorporated into DNA strands
MOA of vinca alkyloids?
bind β-tubulin and block its ability to
polymerize with α-tubulin into microtubules
Vinca alkyloid metabolism?
extensive hepatic metabolism by CYP3A4 requiring dose reduction
for hepatic impairment
Cell cycle phase affected by microtuble targeting agents?
M-Phase
MOA of taxanes (paclitaxel and docetaxel)?
binding of β-tubulin subunit of microtubules and through the PROMOTION of microtubule polymerization
Taxane moiety required for activity?
C13 side chain on 8 member hexane ring
Why does paclitaxel require a 50:50 Etoh to castor oil vehicle?
Lipophilicity
Method of correction for paclitexol lipophilicity?
Abraxane: Paclitexol bound to albumin
Topo-1 inhibitors?
camptothecin analogs
Camptothecin analogs?
topotecan and, irinotecan
Phase of cell cycle affected by topoisomerase inhibitors?
S-phase
Irinotecan (CPT-11) is a prodrug metabolized by hepatic
carboxylesterase to its active form ______.
SN-38
Topoisomerase II inhibitors?
epipodophyllotoxins and anthracycline analogs
Epipodophyllotoxins?
Etopsode
Anthracycline analogs?
doxorubicin, daunorubicin, idarubicin
epirubicin, mitoxantrone
MOA of TOPO-II inhibitors?
- flat 4-RING system intercalates directly with DNA (GC rich) forming a complex with topoisomerase and DNA, causing double-stranded DNA breaks leading to apoptosis. Also produces free radicals that damage DNA.
All anthracyclines are _____toxic?
Cardiotoxic
3 reasons for the cardiotoxicity of anthracyclines?
-quinolone reduction producing superoxide
-no catalase in cardiac tissue
-high cardiac uptake of anthrcyclines
Ion chelator used to reduce the carditoxicity of anthracyclines?
Dexarazoxone
Liposomal anthracycline designed to reduce cardiotoxicity?
Doxorubicin Liposomal (DOXIL)
MOA of all-trans retinoic acid?
-displacement of the repressor of differentiation
-promotion of the degradation of the PML-RARa fusion protein
MOA of arsenic trioxide?
-displacement of the repressor of differentiation
-production of oxygen free radicals
-inhibition of angiogenesis
Effect of ATR and ATRO on PML-RARa?
ATO: Post translational modification of protein
ATRA: Replaces repressor and induces degradation (no post translational modification)
Aromatase inhibitors?
Anastrazole - Breast
Exemestane -Breast
Letrozole - Breast
SERD?
Fulvestrant -Breast
SERMs?
Raloxifene - Breast
Tamoxifen - Breast
MOA of aromatase inhibitors?
inhibits the enzyme aromatase, which is responsible for converting androgens to estrogens
Androgen deprivation therapy?
Goserelin acetate (Zoladex) is an injectable gonadotropin releasing hormone super-agonist (GnRH agonist), also known as a lutenizing hormone releasing hormone (LHRH) agonist. Goserelin acetate is used to suppress production of the sex hormones testosterone and oestrogen), particularly in the treatment of breast and prostate cancer.
GnRH agonists?
Leuprolide acetate and Goserelin acetate
GnRH antagonist?
Abarelix
Androgen receptor blockers?
Flutamide
Nilutamide
Bicalutamide
Oral androgen deprivation agents?
Flutamide
Nilutamide
Bicalutamide
Rituximab?
Anti-CD20 (Lymphoma)
Alemtuzumab?
Anti-CD52 (CLL)
Gemtuzumab-Ozogamicin?
Anti-CD33 + toxin (AML)
Cetuximab?
Anti-EGFR (colorectal cancer)
Panitumumab?
Anti-EGFR (colorectal cancer)
Trastuzumab?
Herceptin: anti-her2/neu (breast cancer)
Bevacizumab?
anti-VEGF (colorectal cancer)
-ximab?
Chimeric
-umab?
Humanized
EGF-EGFR binding promotes?
tumor cell growth and proliferation
Suffix of tyrosine kinase inhibitors?
-nib
Tyrosine kinase inhibitors that inhibit ABL?
Imatinib (CML, ALL)
Dastanib (Leukemia)
Nilotinib (Leukemia)
Tyrosine kinase inhibitors that inhibit EGFR?
Gefitinib (lung)
Erlotinib (lung)
Lapatinib (breast)
Thalidomide complex?
intercalates into DNA in G-C rich regions
Bortezomib MOA?
NF-Kb?