Abstract
Background
In RAS-mutant tumors, combined MEK and autophagy inhibition using chloroquine demonstrated synthetic lethality in preclinical studies. This phase II trial evaluated the safety and activity of the MEK inhibitor binimetinib combined with hydroxychloroquine (HCQ) in patients with advanced KRAS-mutant non-small cell lung cancer (NSCLC).
Methods
Eligibility criteria included KRAS-mutant NSCLC, progression after first-line therapy, ECOG PS 0-1, and adequate end-organ function. Binimetinib 45 mg was administered orally (p.o.) bid with HCQ 400 mg p.o. bid. The primary endpoint was objective response rate (ORR). A Simon’s 2-stage phase II clinical trial design was used, with an α error of 5% and a power β of 80%, anticipating an ORR of 30% to proceed to the 2-stage expansion.
Results
Between April 2021 and January 2022, 9 patients were enrolled to stage I: median age 64 years, 44.4% females, 78% smokers. The best response was stable disease in one patient (11.1%). The median progression free survival (PFS) was 1.9 months, and median overall survival (OS) was 5.3 months. Overall, 5 patients (55.6%) developed a grade 3 adverse event (AE). The most common grade 3 toxicity was rash (33%). Pre-specified criteria for stopping the trial early due to lack of efficacy were met.
Conclusion
The combination of B + HCQ in second- or later-line treatment of patients with advanced KRAS-mutant NSCLC did not show significant antitumor activity. (ClinicalTrials.gov Identifier: NCT04735068).
Keywords: non-small cell lung cancer, KRAS, MEK 1/2 inhibitor, binimetinib, hydroxychloroquine
This phase II trial evaluated the safety and activity of the MEK inhibitor binimetinib combined with hydroxychloroquine in patients with advanced KRAS-mutant non-small cell lung cancer.
Lessons Learned.
The combination of binimetinib and hydroxychloroquine did not exhibit strong antitumor activity in second- or later-line treatment for patients with KRAS-mutant stage IV non-small cell lung cancer.
Discussion
Alterations in KRAS account for the most common mutations in non-squamous non-small cell lung cancer (NSCLC). Despite availability of 2 orally available tyrosine kinase inhibitors (TKIs) that target KRAS G12C-mutant NSCLC, response rates are lower than have been seen with other TKIs. More efficacious targeted treatment approaches are needed. Targeting MAPK signaling has been an attractive strategy to target RAS mutant tumors; however, targeting MAPK alone is complicated due to the emergence of resistance pathways such as autophagy, an adaptive response to metabolic and therapeutic stress. We hypothesized that inhibition of autophagy in combination with MAPK inhibition could be a viable approach to treating KRAS-mutant lung cancer.
Hydroxychloroquine (HCQ) and its derivatives bind and inhibit palmitoyl protein thioesterase 1, a lysosomal enzyme that regulates lysosomal acidification1,2 Inhibition of PPT1 with HCQ deacidifies lysosomes, inhibiting the last step in autophagy. In preclinical models of RAS mutant tumors, combination of a MEK inhibitor with autophagy inhibition using chloroquine or HCQ has demonstrated synergistic antitumor activity.3-5 Binimetinib, an orally bioavailable selective and potent mitogen-activated protein (MAP) kinase kinase (MEK1/2) inhibitor, was approved in 2018 by the US Food and Drug Administration (FDA) for treatment of patients with BRAF-mutant melanoma. We hypothesized that HCQ could enhance the therapeutic efficacy of binimetinib by inhibiting autophagy, serving as an effective treatment strategy for patients with stage IV NSCLC. Using a Simon 2-stage design, this prospective study regimen was to be considered unworthy of further development if the ORR was ≤30%. Nine patients were enrolled in stage I of this clinical trial, without any observed objective responses. Five of 9 patients (56%) developed a grade >3 AE with rash as the most common grade 3 toxicity (33%; Table 1). No grade 4 treatment-related AEs or treatment-related deaths occurred. Our results show that the combination of B and HCQ does not have significant antitumor activity in second-line or beyond treatment of KRAS-mutant stage IV NSCLC.
Table 1.
AEs | n (%) | Grades 1-2, n (%) | Grade 3, n (%) |
---|---|---|---|
Any TRAE | 9 (100) | – | – |
Grades 1-2 TRAE | 9 (100) | – | – |
Grades 3-5 TRAE | 5 (56) | – | – |
Treatment-related SAE | 4 (44) | – | – |
Treatment related to discontinuation | 1 (11) | – | – |
Treatment leading to death | 0 (0) | – | – |
Most common TRAEs (>10% of patients) | |||
Rash | 6 (67) | 3 (33) | 3 (33) |
Diarrhea | 5 (56) | 4 (44) | 1 (11) |
Pruritus | 3 (33) | 3 (33) | 0 (0) |
Fatigue | 3 (33) | 3 (33) | 0 (0) |
Other AEs of interest | |||
Retinal detachment | 1 (11) | 0 (0) | 1 (11) |
Increase in AST | 1 (11) | 0 (0) | 1 (11) |
Increase in ALT | 1 (11) | 1 (11) | 0 (0) |
Abbreviation: AE: adverse events; AST: aspartate aminotransferase; ALT: alanine aminotransferase; SAE: serious adverse event; TRAE: treatment emergent adverse event.
Trial Information | |
---|---|
Disease | Lung cancer—NSCLC |
Stage of disease/treatment | Metastatic/advanced |
Prior therapy | At least one prior regimen |
Type of study | Phase II, single arm |
Primary endpoint | Objective response rate, safety, and tolerability |
Secondary endpoints | Progression-free survival, overall survival |
Investigator’s analysis | Inactive because results did not meet primary endpoint |
Additional details of study design | This single-arm phase II study enrolled patients with cytologically or histologically confirmed stage IV NSCLC with the presence of a non-synonymous mutation in KRAS. The eligibility criteria included age ≥18 years, ECOG PS 0-1, disease progression after at least one prior systemic therapy for metastatic NSCLC, measurable disease based on the RECIST 1.1 criteria, adequate end-organ function, and able to provide written informed consent. Patients with asymptomatic or treated, stable brain metastases were allowed to enrol. All participants provided written informed consent prior to participation. The study was approved by the Institutional Review Board at the University of Pennsylvania and registered at ClinicialTrials.gov (NCT04735068). |
Drug Information | |
---|---|
Generic/working name | Binimetinib |
Company name | Pfizer |
Drug type | Small molecule |
Drug class | MEK 1/2 inhibitor |
Dose | 45 mg per dose |
Route | p.o. |
Schedule of administration | 45 mg 2 times daily over a 28-day treatment course, until disease progression, unacceptable toxic effects, or withdrawal of consent |
Generic/working name | Hydroxychloroquine |
Company name | Commercially available |
Drug type | Immunosuppressant |
Drug class | Immunosuppressant |
Dose | 400 mg per dose |
Route | p.o. |
Schedule of administration | 400 mg by mouth every 12 hours daily over a 28-day treatment course, until disease progression, unacceptable toxic effects, or withdrawal of consent |
Patient Characteristics | |
---|---|
Number of patients, male | 5 |
Number of patients, female | 4 |
Stage | IV |
Age, median (range) | 64 (52-77) years |
Number of prior systemic therapies: median (range) | 3 (2-10) |
Performance status: ECOG | 0-1: 9 2: 0 3: 0 4: 0 |
Cancer types or histologic subtypes | Adenocarcinoma, 9 |
Primary Assessment method: Objective Response Rate | |
---|---|
Number of patients screened | 11 |
Number of patients enrolled | 9 |
Number of patients evaluable for toxicity | 9 |
Number of patients evaluated for efficacy | 9 |
Evaluation method | RECIST 1.1 |
Response assessment, CR | 0 (0%) |
Response assessment, PR | 0 (0%) |
Response assessment, SD | 1 (11.1%) |
Response assessment, PD | 8 (88.9%) |
Median duration assessment, PFS | 1.9 months |
Median duration assessment, OS | 5.3 months |
Assessment, Analysis, and Discussion
Completion | Study completed |
---|---|
Investigator’s assessment | Inactive because results did not meet primary endpoint |
The addition of HCQ to the MEK inhibitor binimetinib did not demonstrate enough activity to justify expansion in a 2-stage phase II trial in patients with KRAS-mutant NSCLC. There were no objective responses, and patients had a short median PFS and OS. Further study of binimetinib plus HCQ in patients with metastatic NSCLC is not warranted based on these results.
Despite being the most mutated gene in nonsquamous NSCLC, KRAS mutations have been historically very difficult to target therapeutically. Recently, 2 targeted agents, sotorasib and adagrasib, were approved by the FDA for patients with KRAS G12C-mutant NSCLC, following progression on first-line therapy for metastatic NSCLC. While these agents are a welcome addition to our armamentarium of targeted therapies, there are currently no viable options for patients with non-G12C KRAS-mutant NSCLC. After treatment with platinum doublet-based regimens and a targeted therapy if appropriate, the only available therapy are agents such as docetaxel, which are associated with very poor response rate and survival outcomes with significant toxicity. There is thus an unmet need for better targeted approaches for patients with KRAS-mutant NSCLC.
Autophagy is a cellular defense mechanism that has evolved to protect cells from adverse environmental conditions, including nutritional deprivation, hypoxia, or therapeutic stress. Activation of autophagy mediates regulated catabolism of cellular organelles, which are encapsulated first in autophagosomes and metabolized when these fuse to lysosomes. Autophagy has been demonstrated to be a resistance mechanism to chemotherapy in several solid tumors. Inhibition of autophagy can be accomplished pharmacologically with HCQ, which inhibits the fusion of the autophagosome to the lysosome. Several clinical trials have demonstrated the safety of this approach in patients with advanced solid tumors. Pharmacokinetic-pharmacodynamic studies have demonstrated that doses of HCQ 400 mg twice daily or higher effectively blocks autophagy when given alone or in combination with other cancer therapies. In addition, in preclinical models, combination of a MEK inhibitor with HCQ has been shown to be synthetically lethal to RAS mutant cancers. The safety and clinical antitumor activity of combining dabrafenib, trametinib (MEK inhibitor), and HCQ has been demonstrated in patients with BRAF-mutant melanoma.6 A randomized trial of this combination therapy is being conducted in patients with BRAFV600-mutant melanoma with elevated LDH and previous immunotherapy. Together, these data provided the rationale for studying a MEK inhibitor in combination with HCQ in KRAS-mutant non-small cell lung cancer.
Nine patients were enrolled onto stage I of this clinical trial. Most patients (77.7%) had received >3 regimens for metastatic disease. Even though all patients had a favorable ECOG PS (0-1) at study entry, most patients had exposure to several lines of previous cytotoxic therapies. Patients on this trial had a variety of molecular alterations, including KRAS G12V, G12D, and G12F.
The combination of HCQ and binimetinib was poorly tolerated. Overall, 5 of 9 patients (55.6%) developed a grade 3 adverse event (AE), with rash being the most common grade 3 toxicity. These toxicities required dose reductions of binimetinib in 2 patients. Most patients (n = 7) received <3 cycles of therapy on trial. The median dose intensity of both drugs was 75%. It is unknown whether longer exposure to this combination might increase the incidence and severity of toxicity. The dose and schedule were predetermined based on other phase I/II trials.7 We did not perform pharmacokinetic testing and cannot exclude the possibility of a drug-drug interaction. These findings suggest that further pharmacokinetic and pharmacodynamic modeling may be required prior to further study so that toxicity can be balanced with efficacy. Expansion to stage II of the trial was not pursued, per protocol, due to lack of activity. Efficacy analysis of the patients treated on the study shows median PFS (1.9 months) and OS (5.3 months). No objective responses were seen.
Our study has some limitations. First, it was a single-arm study with no randomized control design, and hence, selection bias could not be ruled out. Second, we enrolled a heterogenous patient population with different KRAS mutations under the assumption that they would all potentially respond to the studied drug combination. This was based on the preclinical data showing synthetic lethality in RAS mutant cancers regardless of mutation type.
Finally, the dose and schedule were predetermined based on other phase I/II trials, but this might not be the best-suited dose as the next step in patients with NSCLC. Novel autophagy inhibitors such as DCC-3116 (NCT04892017) could be potentially more effective than HCQ, in combination with MEK inhibition in KRAS-mutant NSCLC. In conclusion, the current study showed that the combination of binimetinib combined with HCQ in second- or later-line treatment of advanced KRAS-mutant NSCLC did not exhibit meaningful antitumor activity and is not worthy of further study.
Contributor Information
Charu Aggarwal, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
Alisha P Maity, Department of Internal Medicine, Lankenau Medical Center, Wynnewood, PA, USA.
Joshua M Bauml, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
Qi Long, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
Tomas Aleman, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
Christine Ciunci, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
Christopher D’Avella, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
Melissa Volpe, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
Evan Anderson, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
Lisa McCormick Jones, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
Lova Sun, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
Aditi P Singh, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
Melina E Marmarelis, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
Roger B Cohen, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
Corey J Langer, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
Ravi Amaravadi, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
Funding
This study was sponsored by Pfizer.
Conflict of Interest
Charu Aggarwal reported receiving institutional research funding from AstraZeneca, Genentech, Incyte, Macrogenics, Medimmune, and Merck Sharp & Dohme, and receiving consultation fees from Genentech, Lilly, Celgene Merck, AstraZeneca, Blueprint Genetics, Shionogi, Daiichi Sankyo/AstraZeneca, Regeneron/Sanofi, Eisai, BeiGene, Turning Point, Pfizer, Janssen, and Boehringer Ingelheim. Joshua M. Bauml is currently employed by Janssen R&D, but all work for this study was done as an employee at University of Pennsylvania. Lova Sun reported clinical trial funding (to institution) from Blueprint, Seagen, and IO Biotech, and honoraria/advisory board member for Regeneron, GenMab, Seagen, and MJH Life Sciences. Melina E. Marmarelis reported researching funding (to institution) from Eli Lilly, AstraZeneca, Merck, and Genentech, a consulting role with AstraZeneca, Novocure, Boehringer Ingelheim, Janssen, Takeda, Blueprint Pharmaceuticals, Bristol Myers Squibb, and Ikena, and honorarium from Thermo Fisher, and stock in Gilead Sciences, Portola Pharmaceuticals, Merck, Bluebird Bio, Johnson & Johnson, and Pfizer. Corey J. Langer reported grants from Eli Lilly, Trizell, Merck, Takeda, Inovio, AstraZeneca, Oncocyte, and Boehringer Ingelheim; consulting fees from Boehringer Ingelheim, Novocure, Takeda, Genentech/Roche, AstraZeneca, Merck, Gilead, GSK, Pfizer, Regeneron, Mirati, Sanofi-Aventis, and Heat Biologics; medical writing for Novartis, and DSMB co-chair for Amgen, OncocyteDX, and Veterans Administration. Ravi Amaravadi reported a consulting relationship with Deciphera and Tasca, research funding from Novartis, BMS, Pfizer, and Deciphera, and Scientific Founder of Pinpoint Therapeutics. Alisha P. Maity, Qi Long, Tomas Aleman, Christine Ciunci, Christopher D’ Avella, Melissa Volpe, Evan Anderson, Lisa McCormick Jones, Aditi P. Singh, and Roger B. Cohen indicated no financial relationships.
Data Availability
The data underlying this article will be shared on reasonable request to the corresponding author.
References
- 1. Rebecca VW, Nicastri MC, McLaughlin N, et al. A unified approach to targeting the Lysosome’s degradative and growth signaling roles. Dimeric quinacrines identify new lysosomal target in cancer. Cancer Discov. 2017;7(11):1266-1283. 10.1158/2159-8290.CD-17-0741 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Rebecca VW, Nicastri MC, Fennelly C, et al. PPT1 promotes tumor growth and is the molecular target of chloroquine derivatives in cancer. Cancer Discov. 2019;9(2):220-229. 10.1158/2159-8290.CD-18-0706 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Kinsey CG, Camolotto SA, Boespflug AM, et al. Protective autophagy elicited by RAF→ MEK→ ERK inhibition suggests a treatment strategy for RAS-driven cancers. Nat Med. 2019;25(4):620-627. 10.1038/s41591-019-0367-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Bryant KL, Stalnecker CA, Zeitouni D, et al. Combination of ERK and autophagy inhibition as a treatment approach for pancreatic cancer. Nat Med. 2019;25(4):628-640. 10.1038/s41591-019-0368-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Lee C-S, Lee LC, Yuan TL, et al. MAP kinase and autophagy pathways cooperate to maintain RAS mutant cancer cell survival. Proc Natl Acad Sci USA. 2019;116(10):4508-4517. 10.1073/pnas.1817494116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Mehnert JM, Mitchell TC, Huang AC, et al. BAMM (BRAF Autophagy and MEK Inhibition in Melanoma): a phase I/II trial of dabrafenib, trametinib, and hydroxychloroquine in advanced BRAFV600-mutant melanoma. The BAMM trial in BRAF-mutant melanoma. Clin Cancer Res. 2022;28(6):1098-1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Rangwala R, Leone R, Chang YC, et al. Phase I trial of hydroxychloroquine with dose-intense temozolomide in patients with advanced solid tumors and melanoma. Autophagy. 2014;10(8):1369-1379. 10.4161/auto.29118 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The data underlying this article will be shared on reasonable request to the corresponding author.