Sweatman - Lung Cancer Rx Flashcards

1
Q

Why is the division between NSCLC and SCLC no longer used?

A
  • Histologic subtype of NSCLC now has to be reported due to advent of targeted therapy
  • EGFR mutations are extremely rare in large cell, small cell, and pure squamous carcinomas
    1. Evident in significant # of adenocarcinomas
  • Bevacizumab associated w/high risk of bleeding in squamous lung cancers -> only approved for use in non-squamous NSCLC
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

About what % of NSCLC are squamous vs. adenocarcinoma?

A
  • 50% adenocarcinoma
  • 30% squamous
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

How does the EGF receptor work? How does it “malfunction?”

A
  • Normal cell (A): ligand-induced receptor activation leads to receptor dimerization and trans-phosphorylation of 2 tyrosine residues
    1. Transduced to nucleus via signalling, and nuclear transcription can lead to cell growth, proliferation, and avoidance of apoptosis
  • Cancer cells (B): proliferative pathway gains func via 1) receptor amplification, 2) mutation, or 3) trans-location
    1. Chromosome rearrangement can lead to: the transcriptional activation of proto-oncogenes or creation of fusion genes encoding chimeric proteins w/transforming properties
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

How do TKI’s INH EGFR? What are the 4 NSCLC resistance mechs?

A
  • TKI INH the phosphorylation step, and absence of proliferative signal leads to apoptosis -> this should lead to a clinical anti-tumor response
  • Resistance mechs (see attached image):

C. Drug binding site (T790M) mutation

D. Amplified MET (hepatocyte growth factor receptor, HGFR) phosphorylates ERBB3 (circumventing drug presence)

E. HGF activates P13K/Akt via MET, independent of ERBB3 and EGFR

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

What is an anti-apoptotic IC mech of resistance to TKI’s?

A
  • Polymorphisms in apoptosis genes: downstream of EGFR
    1. Example: BIM polymorphism -> absence of pro-apoptotic BH3 domain in 15% of East Asians (predictive of significantly shorter survival than patients with “normal” BIM)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What is a pro-proliferative IC mech of resistance to TKI’s?

A
  • KRAS and BRAF muts can render anti-EGFR drugs (Mab’s or TKI’s) ineffective due to proliferative path constitutively active DOWNSTREAM of drug-induced blockade
  • Genotyping for mutation can be a deciding factor NOT to treat with Mab, e.g., Cetuximab
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What is the EML4-ALK translocation?

A
  • Family of abnormal (pro-malignant) fusion genes
    1. Echinoderm microtubule-associated protein-like 4 (EML4) and anaplastic lymphoma kinase (ALK) genes
    2. Activates the MEK/ERK pathway and cell proliferation (drug for it now)
  • 2-7% of NSCLC -> more prevalent in non-smokers, pts w/hx of light smoking, & pts w/adenocarcinomas
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Why can blocking VEGF be helpful? Problematic?

A
  • Proliferating tumors need blood supply, which is maintained by cells sensing oxygenation via HIF-1, and the release of VEGF
  • Potential downsides:
    1. Reduce distribution of concurrent chemo
    2. Induce proliferation of more aggressive cells (green in image) that have capacity to spread to other organs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Why is human variation so critical when it comes to anti-cancer drugs, like TKI’s?

A
  • Metabolism may be influenced by several host factors, including bioavailability and host pharmaco-genetics
  • Variation in the germline constitution of individual patients can have significant impact on response to tx and degree of toxicity
  • By inhibiting their target kinases, these agents extinguish crucial downstream signalling pathways involved in normal cell growth
  • Host tissues can also influence efficacy of kinase inhibitors by secreting factors, e.g., HGF, in a paracrine fashion
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Which mutations are more common in non-smokers?

A
  • EGFR (45%)
  • EML4-ALK
  • HER2
  • hMSH2 (40%)
  • Not only smokers get lung cancer
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Why is genetic testing of adenocarcinomas so important?

A
  • Actionable mutation in 60% of patients
  • Data used to either:
    1. Treat according to guidelines based on EGFR results (KRAS and ALK too)
    2. Offer enrollment in clinical trials targeting other ID’d mutations (BRAF, MET)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What are the existing genetic testing guidelines?

A
  • Routine testing recommended for EGFR muts and ALK rearrangements in ALL ADENOCARCINOMAS
    1. Not recommended in pure squamous cell tumors due to very low diagnostic yield
  • DNA sequencing is the method used in most EGFR studies -> discordance of up to 28% b/t 1o tumors and distant metastases
  • Fluorescence in situ hybridization (FISH) preferred and FDA-approved choice for ALK gene rearrangement testin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What is the current recommendation for preventive scans for smokers?

A
  • Smokers (55-80 y/o) with >30-pack history and those who have quit in the last 15 years should get low-dose CT scans of lungs to look for possible tumors
    1. 2 previous reviews found insufficient evidence to support annual screenings
  • Small risk of radiation exposure from low-dose scans outweighed by benefits of detecting abnormal growths early and intervening w/tx
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What are the standard tx plans for SCLC and NSCLC?

A
  • SCLC: metastasis early, so radiation/chemo the only option
  • NSCLC: sx resection if no metastasis (early stage)
    1. Chemo/radiation used in adjuvant, neo-adjuvant or maintenance role to:
    a) reduce bulk of the tumor (facilitating sx) and eradicate early micrometastases OR
    b) prevent growth or metastasis
    2. Pre-op chemo may delay potentially curative sx -> metastasize very early, most commonly spreading to adrenals, liver, brain, and bone
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What are the standard tx options for SCLC?

A
  • Etoposide + Cisplatin or Carboplatin

OR

  • Cisplatin + Irinotecan
  • Cisplatin + Etoposide + Ifosfamide
  • Cyclophosphamide + Doxorubicin + Etoposide
  • ”” + Vincristine
  • Cyclo + Doxo + Vincristine
  • Cyclo + Etoposide + Vincristine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What are the standard tx options for NSCLC?

A
  • Cisplatin AND Paclitaxel, Gemcitabine, Docetaxel, Vinorelbine, Irinotecan, or Pemetrexed (commonly a taxane)
  • Pemetrexed maintenance: for pts w/stable or responding disease after 4 cycles of nonpemetrexed-platinum combo therapy)
  • Based on appropriate genetic markers, TKI’s and VEGF inhibitors may also be used:
    1. EGFR TKI’s for pts w/EGFR+ genetic test
    2. Bevacizumab (pts w/non-squamous histo, no brain metastases, and no hemoptysis)
    3. Erlotinib: first-line tx for locally advanced or metastatic NSCLC after failure of at least 1 prior chemo regimen, or for maintenance tx of “” that has not progressed after 4 cycles of platinum-based first-line chemo
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What is the MOA for Cisplatin and Carboplatin?

A

Form DNA intrastrand crosslinks and adducts

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

What is the MOA for Cyclophosphamide?

A

Pro-drug of active alkylating moiety

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

What is the MOA for Docetaxel?

A

Microtubule stabilizer inhibiting depolymerization

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

What is the MOA for Doxorubicin?

A

Intercalator, free radical generator, topo II INH

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

What is the MOA for Etoposide/VP-16?

A

DNA-topo II complex stabilizer

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

What is the MOA for Gemcitabine?

A

DNA polymerase inhibitor via incorporation of triphosphate form during DNA synthesis

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

What is the MOA for Ifosfamide?

A

Intra- and interstrand crosslinker

24
Q

What is the MOA for Irinotecan?

A

DNA-topo I complex stabilizer

25
Q

What is the MOA for Paclitaxel?

A

Microtubule stabilizer inhibiting depolymerization

26
Q

What is the MOA for Pemetrexed?

A

DHFR inhibitor

27
Q

What is the MOA for Topotecan?

A

DNA-topo I complex stabilizer

28
Q

What is the MOA for Vincristine/Vinorelbine?

A

Microtubule inhibitor; tubules disintegrate into spiral aggregates/protofilaments

29
Q

What is a big clinical issue, and often the dose-limiting toxicity for most of the anti-cancer drugs?

A

Myelosuppression

30
Q

What are the Carboplatin AE’s?

A
  • Allergic (platinum) reactions
  • Dose-related myelosuppression; cumulative anemia
  • Dose-related N/V
  • Blood chemistry dyscrasia, INC hepatic enzymes, BUN, and creatinine
31
Q

What are the Cisplatin AE’s?

A
  • Allergic (platinum) reactions
  • Dose-related, severe nephrotoxicity, myelosuppression and N/V
  • Significant ototoxicity (tinnitus and occasionally deafness) reported in children
32
Q

What are the Cyclophosphamide AE’s?

A
  • Blood dyscrasias -> anemia, infection
  • Renal compromise, hemorrhagic cystitis (Mesna is protective), N/V, rashes
  • Amenorrhea, infertility
  • Monitor for 2o malignancies
  • Pulmonary fibrosis
33
Q

What are the Docetaxel AE’s?

A
  • INC mortality in NSCLC
  • Edema (give steroids)
  • Contraindicated with INC bilirubin/ALK/phos, AST/ALT
  • Dose-limiting neutropenia
  • Sensory neuropathy
34
Q

What are the Doxorubicin AE’s?

A
  • Myelosuppression, CHF (cumulative dose exposure manner: 450 mg/sq m max cumulative dose), hepatic disease
  • 2o malignancies
  • Extravasational necrosis
  • N/V
35
Q

What are the Etoposide/VP-16 AE’s?

A
  • Myelosuppresion, infection
  • Dose-limiting hematologic toxicity
  • N/V, diarrhea
  • Alopecia
36
Q

What are the Gemcitabine AE’s?

A
  • Myelosuppression, infection
  • Arthralgia, drowsiness, fatigue
  • N/V
  • Alopecia
  • Sensory peripheral neuropathy (10%)
37
Q

What are the Ifosfamide AE’s?

A
  • Alopecia
  • N/V
  • Blood dyscrasia -> infection
  • Neurotoxicity
  • Hematuria renal failure (<10%)
38
Q

What are the Irinotecan AE’s?

A
  • Myelosuppression
  • Diarrhea
  • Asthenia
  • Fever, pain
  • Weight loss
39
Q

What are the Paclitaxel AE’s?

A
  • Taxane hypersensitivity
  • Myelosuppression
  • Myalgia/arthralgias
40
Q

What are the Pemetrexed AE’s?

A
  • Myelosuppression and GI toxicities, esp. when combined with Cisplatin vs. NSCLC
  • Elevated LFT’s and serum creatinine
41
Q

What are the Topotecan AE’s?

A
  • Myelosuppression and GI toxicities
  • Hyperbilirubinemia
42
Q

What are the Vinblastine/Vinorelbine AE’s?

A
  • Myelosuppression
  • Neuropathic toxicity (less so w/Vinorelbine)
  • Neutropenia (Vinorelbine)
  • Intrathecal admin of Vinca alkaloids is fatal
43
Q

What are the TKI’s approved for treating NSCLC?

A
  • ALK
    1. Crizotinib: + HGFR
    2. Ceritinib: ALK intolerant to Crizotinib
  • EGFR
    1. Afatinib
    2. Erlotinib
    3. Gefitinib
44
Q

What are some of the pitfalls associated with the oral route of administration of the TKI’s?

A
  • Food may impact bioavailability: reflected in dosing instructions
  • Extensive hepatic metabolism (except Afatinib), and predominantly fecal elimination
  • Substrates for CYP3A4/5 and P-gp (except Crizotinib)
    1. Other isoforms vary by drug
  • Inhibitors of CYP3A, and to varying degrees, other isoforms
  • Drug-drug interactions are possible
45
Q

What are some of the (un)common TKI side effects?

A
  • Common:
    1. EGFR: diarrhea, N/V, dermatologic, elevated hepatic enzymes, opthalmic (except Afatinib)
    1. ALK: constipation, diarrhea, N/V
  • Uncommon:
    1. Gefitinib - renal
    2. Ceritinib - fatigue, DEC hemoglobin, hyperglycemia (but no opthalmic, unlike Crizotinib)
46
Q

What are the rare AE’s with the TKI’s? How should pts be counseled about these?

A
  • Bradyarrhythmia and/or QT prolongation
  • Severe rash and Steven Johnson Syndrome (SJS)
  • GI hemorrhage or perforation
  • Hepatotoxicity and hepatic failure
  • Interstitial lung disease, pneumonia, PE
  • Corneal ulceration, perforation, keratitis
  • Counseling:
    1. Avoid pregnancy -> use appropriate contraception
    2. Immediately report new CV or pulm sxs
    3. Immediatley report bullous, blistering, or exfolative dermatologic symptoms
47
Q

What do you see here?

A
  • EGFR-induced rash: common and potentially severe AE
  • May necessitate specific txs with steroids, antimicrobials, and oatmeal lotions

-

48
Q

What should you do if your lung cancer pt on TKI’s presents with a rash?

A
49
Q

What 2 things do these graphs tell you?

A
  • Erlotinib produces a significant survival effect, especially in the short-term
  • This effect is dependent on the molecular target, in this case, EGFR overexpression by the tumor
50
Q

How is TKI resistance related to ATP affinity?

A
  • Muts in TK domain of EGFR can have profound effect on drug sensitivity (ATP binding site highly conserved among TKI’s)
    1. About 10-25% of NSCLS from E. Asia have somatic activating muts -> in-frame deletions in exon 19 (point mutation that substitutes Leu-858 w/arginine; L858R)

A. Compromises ATP affinity and INC initial sensitivity to TKI’s: erlotinib/afatinib (opens the therapeutic window)

  1. Secondary mut (T790M) closes window -> restores ATP affinity to wild-type levels)

A. Detection of this mut in naive pts indicates resistance; may reflect outgrowth of resistant clones (can be in pts who’ve never had TKI’s)

51
Q

What is the theoretical resistance mutation to Crizotinib?

A
  • G2032R ROS1: has been shown to reduce drug activity in in vitro kinase activity assays
  • Emergence of resistance most likely reflects an outgrowth of pre-existing tumor clones with inherent resistance at this site
52
Q

What is PD-1?

A
  • Programmed cell death protein: one of the co-stimulatory signals that serve to modulate the signal (signal 1) in T-cells after initial binding of an Ag-presenting cell (APC)
    1. Binding of PD-1 receptors by PDL1 or PDL2 leads to an INH or (-) regulatory impact on signal 1
    2. Primary role is to regulate inflammatory responses in tissues by effector T cells recognizing Ag in peripheral tissues
    3. Activated T cells upregulate PD-1 and continue to express it in tissues -> inflammatory signals in tissues induce expression of PD1 ligands that downregulate the activity of T cells, limiting colateral damage in response to a microorganism in that tissue
53
Q

Nivolumab

A
  • Very recent approval -> metastatic disease after failure of platinum therapy
  • Toxicity profile limited (for now) -> common AE’s similar to those for TKI’s
    1. Pruritis/rash, hyperkalemia/hyponatremia, DEC appetite, constipation, N/V, elevated LFT’s, cough, musculoskeletal pain, respiratory infection, fatigue
  • Less common AE’s: endocrine dysfunction, immune-mediated organ destruction
  • Pt counseling: use reliable contraception, promptly report new sxs of dermatologic, hepatic, renal, or endocrine dysfunction
54
Q

Bevacizumab

A
  • Humanized Ab given IV; binds VEGF and prevents receptor activation
  • Common AE’s similar to those of TKI’s:
    1. HTN, hand-foot, endocrine, GI, or neuro dysfunction, hemorrhage, infections, proteinuria, fatigue
  • Less common effects: arterial thromboembolism, CHF, DVT, cerebral artery occlusion, pulm HTN
    1. Impaired wound healing
    2. Fistula formation in tracheoesophagus/GI tract, liver, renal pelvis, F genital tract, bronchopleural tissue
55
Q

What is the downside of Bevacizumab?

A
  • INH blocks endo cell regeneration leading to underlying matrix exposure, and thrombosis/hemorrhage
  • INH also promotes non-physiologic apoptosis of endo cells and DEC deposition of sub-endothelial matrix, making vasculature more susceptible to bleeding
  • Life-threatening, severe bleeding is possible
  • Gastric perforation and impaired wound healing may also contribute to hemorrhage in some cases
56
Q

Can you use Bevacizumab in the tx of squamous NSCLC?

A
  • NO -> contraindicated in these patients due to increased risk of bleeding bc located near major blood vessels, and necrose and cavitate
  • Baseline cavitation was the main risk factor of high-grade bleeding for patients with NSCLC
  • Drug causes central necrosis and enlarged tumor cavitation in these patients -> this combo w/immature blood vessels in the cavity leads to INC risk of bleeding
57
Q

How does clinical staging of lung cancer affect genetic testing?

A