Unit 1 - Molecular Targeted Therapy Flashcards

1
Q

explain what preclinical studies are

A

new drug is tested on normal and cancer cell lines in a laboratory setting
-lab investigation involves animal testing for efficacy and toxicity

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

explain what an investigational new drug (IND) application is

A

new drug sponsor files an IND application with FDA for clinical testing in humans
-if FDA approves application, clinical trials start

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

explain what phase I trials are

A

determine safe and appropriate dose for subsequent studies

  • have 10-30 pts with different malignancies that failed standard treatment, and life expectancy of at least 1-2 months with functioning organs capable of drug metabolism and excretion
  • highest dose with acceptable toxicity is recommended for PII
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

explain what phase II trials are

A

determine effectiveness and side effects of new drug

  • have up to 100 pts with same type of malignancy for which no effective treatment is available and/or are most likely to respond to therapy, with life expectancy at least 3 mo and functioning organs with limited number of prior treatments
  • patients are closely monitored and immediately removed from treatment if condition worsens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

explain what phase III trials are

A

evaluates effectiveness of new drug, and compares it to best available standard treatment
-large and long-term trials with 100-1000s of patients whose eligibility depends on disease setage, first time treatment, status, goals, etc.

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

explain what a new drug application is

A

drug sponsor files NDA or BLA (biologics license application) with FDA
-sponsor submits phase III trial data indicating new drug is safe and superior to standard treatment

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

explain what FDA approval is

A

if satisfied with phase III results, FDA approves new drug, which can take ~1.5 years
-after approval, drug can be marketed to public under a label that indicates dosage, safety, indications, and side effects

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

explain what phase IV trails are

A

determine long-term safety and effectivfeness of new drug

-since III isn’t long enough to see long-term problems

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

mechanism of action of Imatinib (Gleevec)

A

selective activity against Abl tyrosine kinase of CML
-binds to Bcr-Abl at same site where ATP binds, thus blocking Bcr-Abl’s ability to phosphorylate and activate proteins involved in malignant transformation

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

toxicity of Imatinib (Geevec)

A

minimal side effects, including nausea, vomiting, fluid retention, muscle cramps, and arthralgia
-myelosuppression in 29% of patients

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

why was there intrinsic resistance to Imatinib (Gleevec)?

A

if persistent Bcr-Abl kinase activity, could be due to:

  • mutations in Bcr-Abl kinase, making it insensitive to drug
  • drug unable to reach target b/c enhanced binding to other proteins in circulation and/or drug efflux
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

why was there relapse after initial responses to Imatinib?

A

mostly involves reactivation of Bcr-Abl kinase
-if mutations in Abl kinase, mutant kinase becomes less sensitive to drug
-unknown mechanism behind patients with Bcr-Abl amplification
, or persistent inhibition of Bcr-Abl kinase

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

what are Nilotinib and Dasatinib?

A

2nd generation TKIs

  • Nilotinib: better fit in ABL pocket (20-30x potency) and more effective if Imatinib-resistant
  • Dasatinib: side effect is pulmonary arterial HTN (PAH)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

explain what GISTs are

A

gastrointestinal stromal tumors; most common mesenchymal malignancy of GIT (~1%)

  • benign and malignant, but all GISTs are potentially malignant
  • commonly stomach > SI > esophagus > colon > rectum
  • don’t involve lymph nodes, but metastasize to liver
  • generally in older adults
  • 90% are KIT (CD117)+, 5% have PDGFRA mutations (both are TK receptors)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

how does imatinib treat GIST?

A

inhibits KIT and PDGFRA at 400 mg/day (although up to 800 mg/d is safe, no better effect)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

explain what the Ph+ in CML is

A

Philadelphia Xm in >90% of cases, that’s a reciprocal translocation between Xm 9 and 22
-t(9,22) fuses BCR gene on Xm 22 with ABL gene from Xm 9, creating famous Bcr-Abl fusion protein

17
Q

explain what the PML-RARalpha in APL is

A

reciprocal translocation between Xm 15 and 17 in 98% of cases
-fuses retinoic acid receptor alpha gene with promyelocytic leukemia gene to make PML-RAR-alpha

18
Q

explain the mechanism of action of retinoic acid in APL

A

induces terminal differentiation of malignant cells that subsequently undergo natural apoptosis

  • in normal cells, RA binds to its receptors including RAR-alpha, to induce expression of genes involved in myeloid cell differentiation
  • at pharmacological concentration, RAR-alpha is activated via unknown mechanisms to induce expression of genes involved in myeloid cell differentiation
19
Q

what is the toxicity of retinoic acid for APL?

A

if used alone, 1/3 of patients get increased WBC (leukocyte activation/retinoic acid syndrome)
-fever, respiratory distress, weight gain, pleural/pericardial effusion, renal failure

-if concurrent uses of chemo with retinoic acid plus corticosteroids, blocks LAS, but still has dryness of skin/lips, nausea, headache, arthralgia, bone pain

20
Q

explain what the EGFR family members are

A

epidermal growth factor receptors in cancer development and progression

  • ErbB1 (EGFR, HER1) is activated by EGF, TGF-alpha
  • ErbB2 (HER2/neu) has no known ligand, but is the master coordinator that shares the ErbB ligands
  • ErbB3 (HER3) is activated by neuregulin
  • ERbB4 (HER4) is activated by neuregulin
21
Q

in which cancers in ErB1 and 2 overexpressed

A

1: gastric, breast, prostate, bladder, ovarian, colorectal, non-small cell lung, glioblastoma
2: breast, ovarian, gastric

22
Q

what is trastuzumab’s target? mechanism?

A

against Erb-B2 (HER-2/neu) growth factor receptor

  • seems to prevent transduction of proliferation and survival signals
  • induces cytostatic growth inhibitory effects against ErbB2 overexpressing cells, due to Ab-induced downregulation and degradation of receptor
  • monotherapy is modest response, but better if combined
23
Q

what is trastuzumab’s toxicity?

A

hypersensitivity reaction (even if humanized version)

  • ventricular dysfunction and CHF
  • can enhance cardiac toxicity of doxorubicin, so give in combo with taxanes
24
Q

whats the mechanism of cetuximab and against what? its toxicity?

A

anti-ErbB1 monoclonal Ab

  • competes for ligands to inhibit ErbB1 TK activity and growth promoting/survival signals
  • works better with cisplatin and other standard chemotherapy agents
  • causes pulmonary embolism and allergies
25
Q

what is rituximab and its mechanism?

A

treatment for non-Hodgkin’s lymphoma

  • binds to CD20 Ag, which is TMP present on all B-cells
  • -eliminates CD20+ follicular lymphoma cells by: direct activation of apoptosis, complement activation, cell-mediated cytotoxicity
26
Q

what are the most common types of cutaneous melanoma?

A
  1. superficial spreading melanoma (70%)
  2. nodular melanoma (15%; common in males >50, but occur at any age)
  3. acral lentiginous melanoma (5%; more common in darker skinned)
  4. Lentigo maligna melanoma (10%; common in middle aged to old)
27
Q

what is the mutation most commonly attributed to cutaneous melanomas?

A

BRAF, which codes a serine threonine kinase

-V–>E most common, then V–>K

28
Q

what drug is used to treat cutaneous melanomas? how does it work?

A

Vemurafenib is a BRAF inhibitor

-blocks constitutively activated pathway and downstream activity, decreasing cell proliferation and survival

29
Q

side effects and contraindications of vemurafenib

A

arthralgia, fatigue, photosensitivity, alopecia, nausea, and diarrhea

  • cutaneous squamous cell carcinoma, keratoacanthoma, or both
  • QT prolongation, thus increased risk of ventricular arrhythmias
  • new cutaneous melanoma

shouldn’t be used if wild type BRAF, electrolyte abnormalities, or long QT syndrome

30
Q

what is Dabrafenib used for? side effects?

A

next generation agent for treatment of cutaneous melanomas

  • higher risk to develop cutaneous squamous cell carcinoma, keratoacanthoma, and melanoma
  • Serious febrile drug reactions (fever associated with hypotension, rigors, chills, dehydration, kidney failure)
  • uveitis, iritis
  • hyperglycemia (needs modification in DM management)
  • may inhibit fertility in males
31
Q

what is Trametinib used for? toxicities?

A

for cutaneous, unresectable or metastatic melanomas

  • has different mech than usual
  • causes cardiomyopathy, retinal pigment epithelial detachment, retinal vein occlusion, interstitial lung disease, serious skin toxicity, and may inhibit fertility in females
  • shouldn’t be given to patients previously treated with BRAF inhibitors
32
Q

what is immunotherapy for melanoma?

A
  1. ipilimumab (Yervoy); inhibits CTLA-4 to stimulate immune system
  2. IL-2