Targeting ALK, ROS1, and BRAF Kinases
2012; Elsevier BV; Volume: 7; Issue: 12 Linguagem: Inglês
10.1097/jto.0b013e31826df05e
ISSN1556-1380
AutoresRobert C. Doebele, D. Ross Camidge,
Tópico(s)Melanoma and MAPK Pathways
ResumoCancer cells demonstrate numerous genetic aberrations. Despite this genetic complexity, it is hypothesized that cancer cells are often addicted to a single oncogenic driver such that inhibition of this single target would lead to cell death.1Weinstein IB Cancer. Addiction to oncogenes–the Achilles heal of cancer.Science. 2002; 297: 63-64Crossref PubMed Scopus (1477) Google Scholar Data collected from patients with adenocarcinoma by the Lung Cancer Mutation Consortium demonstrate a significant number of patients harboring distinct oncogenic drivers, many of which may be amenable to targeted therapies.2Kris MG Giaccone G Davies A et al.Systemic therapy of bronchioloalveolar carcinoma: results of the first IASLC/ASCO consensus conference on bronchioloalveolar carcinoma.J Thorac Oncol. 2006; 1: S32-S36Abstract Full Text Full Text PDF PubMed Google Scholar Two current areas of intense investigation are to match targeted therapies to a growing list of selected oncogene aberrations in patients with non–small-cell lung cancer (NSCLC) and also to understand mechanisms of resistance to these targeted therapies so that resistance can be overcome or potentially delayed with new drugs or drug combinations. The recent approval of crizotinib for anaplastic lymphoma kinase (ALK) positive NSCLC patients ushered in the first Food and Drug Administration companion diagnostic for NSCLC, ALK fluorescence in situ hybridization (FISH). ALK break-apart FISH was, and still is, the diagnostic test used for entry into the completed and ongoing crizotinib studies and remains the only prospectively validated test for this drug. Dr. Garcia described other testing methods that are in development, including immunohistochemistry (IHC) and bright field in situ hybridization (BISH), both of which might have advantages relating to ease of adoption. Whether either would also have an advantage in terms of cost effectiveness is debatable as this will depend on the ultimate cost of a validated assay. IHC takes advantage of the lack of ALK expression in normal lung tissue, or in cancers without an ALK gene rearrangement, by evaluating levels of ALK protein expression. BISH uses a similar strategy to FISH by evaluating the distance between chromagen-labeled probes that are homologous to the 5′ and 3′ ends of the ALK gene to detect evidence of an inversion or translocation involving the ALK gene, but does so without the need for a fluorescence microscope. Although early studies suggest good concordance between these tests and ALK FISH within single centers, the need for both standardized protocols for the multiple commercially available ALK antibodies for IHC and the need for standardized scoring systems for both BISH and IHC were highlighted. Drs. Doebele and Shaw presented data on observed mechanisms of resistance to crizotinib in ALK positive NSCLC patients who underwent biopsy at the time of progression.3Doebele RC Pilling AB Aisner DL et al.Mechanisms of resistance to crizotinib in patients with ALK gene rearranged non-small-cell lung cancer.Clin Cancer Res. 2012; 18: 1472-1482Crossref PubMed Scopus (918) Google Scholar,4Katayama R Shaw AT Khan TM et al.Mechanisms of acquired crizotinib resistance in ALK-rearranged lung Cancers.Sci Transl Med. 2012; 4: 120ra17Crossref PubMed Scopus (1033) Google Scholar Both groups observed a diverse array of ALK kinase domain mutations. The diversity of mutations is reminiscent of those observed in BCR-ABL after resistance to imatinib, rather than the dominant T790M mutation observed in epidermal growth factor receptor (EGFR) mutant patients after EGFR kinase inhibitor resistance. ALK resistance mutations were observed in 20% to 40% of patients. Copy number gain through amplification of the ALK gene fusion was observed in 5% to 20% of patients. The University of Colorado series observed 36% of patients with the presence of an EGFR or v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) activating mutation and/or the absence of an ALK gene rearrangement in the resistance biopsy, suggesting the emergence of a clonally distinct population of cells that were no longer solely dependent on ALK signaling. The Massachusetts General Hospital series observed increased phosphorylated EGFR in the absence of an activating mutation in a subset of patients. Previously, some cell lines derived from crizotinib-naive ALK+ patients have also demonstrated increased EGFR and/or v-erb-b2 erythroblastic leukemia viral oncogene homolog 2 (HER2) signaling in the absence of activating mutations, with a combination of agents directed against both ALK and EGFR/HER2 signaling required to achieve growth inhibition.4Katayama R Shaw AT Khan TM et al.Mechanisms of acquired crizotinib resistance in ALK-rearranged lung Cancers.Sci Transl Med. 2012; 4: 120ra17Crossref PubMed Scopus (1033) Google Scholar,5Koivunen JP Mermel C Zejnullahu K et al.EML4-ALK fusion gene and efficacy of an ALK kinase inhibitor in lung cancer.Clin Cancer Res. 2008; 14: 4275-4283Crossref PubMed Scopus (833) Google Scholar In addition, from the Massachusetts General Hospital series, v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog (KIT) gene amplification was observed as a mechanism of resistance in two patients. Collectively, these data suggest that resistance to crizotinib in some patients may emerge through total or partial reliance on other oncogenic drivers, and the primary significance of this is that these patients would not be expected to benefit from monotherapy with a more potent ALK inhibitor. In contrast, patients with ALK kinase domain resistance mutations and copy number gain of the ALK fusion gene are likely to still retain oncogene addiction to ALK signaling. Thus, these patients may be much more likely to benefit from a more potent, next-generation, ALK kinase inhibitor. Drs. Shaw, Gettinger, and Gandhi presented preclinical data on three such ALK inhibitors: LDK378 (Novartis, Basel, Switzerland), AP26113 (Ariad, Cambridge, MA), and CH5424802 (Chugai), respectively. In xenograft models, all three drugs demonstrated activity against unmutated EML4-ALK whereas LDK378, and AP26113 also demonstrated activity against xenografts bearing the known C1156Y and L1196M crizotinib-resistance mutations, respectively. Interestingly, AP26113 demonstrates activity against mutant EGFR, which could be applicable in patients who demonstrate this mechanism of resistance, although the IC50s (median inhibition concentration) associated with EGFR inhibition were significantly higher than for ALK inhibition. Currently, all three compounds are being evaluated in early-phase studies in both crizotinib-naive and crizotinib-resistant patients. AP26113 is also being evaluated in cancers with a c-ros oncogene 1, receptor tyrosine kinase (ROS1) gene rearrangement (see below). Dr. Camidge described a phase I study combining crizotinib with dacomitinib (PF-299804), Pfizer's irreversible pan-HER inhibitor. This drug combination was initially developed to address the common mechanisms of acquired resistance in EGFR mutant NSCLC (T790M and met proto-oncogene (MET) gene amplification), however, it may also have a role in crizotinib resistance occurring through increased EGFR and/or HER2 signaling and accrual to a dedicated ALK+ crizotinib-resistant cohort is being considered. Given the diversity of resistance mechanisms to crizotinib and their potential impact on subsequent treatment strategies directly and solely against ALK, it will be critical to evaluate the mechanism of crizotinib resistance in the patients treated within all these studies. ROS1 is a receptor tyrosine kinase that is homologous to ALK. Similar to the ALK FISH break-apart assay, the break-apart FISH assays, according to Dr. Garcia, were meant to evaluate the deletions, inversions, and translocations that can fuse the 3′ region encoding the kinase domain of the ROS1 oncogene to a variety of 5′ fusion partners including CD74, EZR, GOPC (FIG), LRIG3, SL34A2, SDC4, and TPM3.6Bergethon K Shaw AT Ou SH et al.ROS1 rearrangements define a unique molecular class of lung cancers.J Clin Oncol. 2012; 30: 863-870Crossref PubMed Scopus (1265) Google Scholar,7Takeuchi K Soda M Togashi Y et al.RET, ROS1 and ALK fusions in lung cancer.Nat Med. 2012; 18: 378-381Crossref PubMed Scopus (1037) Google Scholar The incidence across multiple studies suggests that ROS1 gene fusions occur in slightly more than 1% of NSCLC. In vitro data suggest that ROS1 can transform cells and that inhibition with the ALK inhibitors, crizotinib, or TAE684, blocks cell proliferation and induces cell-cycle arrest in G1 via inhibition of downstream signaling cascades through SHP2, AKT, and ERK.7Takeuchi K Soda M Togashi Y et al.RET, ROS1 and ALK fusions in lung cancer.Nat Med. 2012; 18: 378-381Crossref PubMed Scopus (1037) Google Scholar,8Davies KD Le AT Theodoro MF et al.Identifying and Targeting ROS1 Gene Fusions in Non-Small Cell Lung Cancer.Clin Cancer Res. 2012; 18: 4570-4579Crossref PubMed Scopus (355) Google Scholar Treatment with crizotinib in an expanded cohort of the Pfizer phase I trial of PF-02341066 (crizotinib) induced tumor shrinkage in two patients with ROS1 gene rearrangements, one with an syndecan 4 (SDC4–ROS1) rearrangement, and the other unknown, suggesting that these gene rearrangements may be a suitable target for inhibitors such as crizotinib.6Bergethon K Shaw AT Ou SH et al.ROS1 rearrangements define a unique molecular class of lung cancers.J Clin Oncol. 2012; 30: 863-870Crossref PubMed Scopus (1265) Google Scholar,8Davies KD Le AT Theodoro MF et al.Identifying and Targeting ROS1 Gene Fusions in Non-Small Cell Lung Cancer.Clin Cancer Res. 2012; 18: 4570-4579Crossref PubMed Scopus (355) Google Scholar The v-raf murine sarcoma viral oncogene homolog B1 (BRAF) encodes a nonreceptor serine/threonine kinase that can activate the MAPK pathway. Activating mutations in BRAF occur in approximately 3% of NSCLC patients, much lower than that observed in melanoma, where investigations have recently led to the Food and Drug Administration approval of vemurafenib in this disease.9Paik PK Arcila ME Fara M et al.Clinical characteristics of patients with lung adenocarcinomas harboring BRAF mutations.J Clin Oncol. 2011; 29: 2046-2051Crossref PubMed Scopus (548) Google Scholar,10Chapman PB Hauschild A Robert C BRIM-3 Study Group et al.Improved survival with vemurafenib in melanoma with BRAF V600E mutation.N Engl J Med. 2011; 364: 2507-2516Crossref PubMed Scopus (6115) Google Scholar The most common mutation, V600E, is seen in approximately 50% of NSCLC patients with a BRAF mutation, whereas the other less-common non-V600E mutations (G469A and D594G) comprise the other half. Drs. Miller and Riely reported on preclinical and clinical activity of dabrafenib (GSK2118436; Glaxo-SmithKline, Brentford, Middlesex, United Kingdom) and vemurafenib (PLX4032; Genentech, South San Francisco, CA), respectively. Both drugs are being investigated in a number of BRAF mutant malignancies. One significant safety concern with this class of drug is the induction of squamous cell carcinomas of the skin in less than 20% of patients, which typically occur within weeks to months of initiation of the drug and are treated successfully with curative-intent curettage.10Chapman PB Hauschild A Robert C BRIM-3 Study Group et al.Improved survival with vemurafenib in melanoma with BRAF V600E mutation.N Engl J Med. 2011; 364: 2507-2516Crossref PubMed Scopus (6115) Google Scholar One interesting mechanism of intrinsic resistance that has come to light in BRAF mutation-positive colorectal cancer is the activation of EGFR via inhibition of BRAF, which inhibits a phosphatase that typically inactivates EGFR.11Prahallad A Sun C Huang S et al.Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR.Nature. 2012; 483: 100-103Crossref PubMed Scopus (1480) Google Scholar This finding highlights a potential pitfall of transferring a targeted therapy from one cancer type to another. As the repertoire of targetable genetic abnormalities increase, we will need to develop mechanisms that are both practical and reliable to identify patients with relatively rare oncogenes. The broad range of crizotinib-resistance mechanisms observed in ALK+ NSCLC suggests that diversity will re-emerge as a major issue even from initially largely molecular uniform tumors in the acquired-resistance setting. Whether therapies directed against resistance mechanisms should be employed only at the time of resistance, or earlier to delay the emergence of resistance, will need to be addressed over the next few years. Factors influencing the choice between these two strategies will include the tolerability of the treatment regimen and whether technology looking, for example, for low levels of resistance mechanisms in treatment-naive patients, will evolve to allow us to accurately predict which patient is more likely to employ a specific mechanism of resistance before it becomes clinically apparent.
Referência(s)