Repurposing an HIV drug to improve efficacy of targeted therapy in melanoma
Commentary

Repurposing an HIV drug to improve efficacy of targeted therapy in melanoma

Lawrence W. Wu, Gao Zhang, Meenhard Herlyn

Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, Pennsylvania, USA

Correspondence to: Meenhard Herlyn. The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA. Email: herlynm@wistar.org.

Comment on: Smith MP, Brunton H, Rowling EJ, et al. Inhibiting Drivers of Non-mutational Drug Tolerance Is a Salvage Strategy for Targeted Melanoma Therapy. Cancer Cell 2016;29:270-84.


Submitted May 11, 2016. Accepted for publication May 19, 2016.

doi: 10.21037/tcr.2016.05.28


Targeted mitogen-activated protein kinase inhibitor (MAPKi) therapies have had limited efficacy in patients with v-Raf murine sarcoma viral oncogene homolog B (BRAF) -mutant, unresectable or metastatic melanomas and tumor relapse is almost inevitable (1). There has been a great deal of studies dissecting heterogeneous molecular mechanisms of acquired resistance to mutant BRAF-targeted therapies. For example, up-regulation of mitochondrial biogenesis and altered tumor bioenergetics (2), increased phosphorylation of protein kinase B (AKT) (3), and selection for subpopulations expressing epidermal growth factor receptor (EGFR) (4) are mechanisms responsible for acquired resistance. Some approaches to overcome acquired drug resistance are combining MAPKi with immune checkpoint blockade inhibitor targeting programmed cell death protein 1 (PD-1) (5), targeting both the MAPK and phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/AKT pathway (6), and targeting mitochondrial biogenesis through inhibition of tumor necrosis factor receptor-associated protein 1 (TRAP1) (2). However, much work needs to be done in investigating and therapeutically preventing the emergence of the initial intrinsic resistance to MAPKi.

Several studies have implicated microphthalmia-associated transcription factor (MITF) as a key driver of intrinsic drug resistance. Drug-sensitivity to MAPKi is correlated with expression and activity of MITF and inversely correlates with nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and AXL receptor tyrosine kinase (AXL) expression (7). A MITF-low/AXL-high/drug-resistance phenotype is common in BRAF- and neuroblastoma RAS viral oncogene homolog (NRAS)-mutant melanoma cell lines (8). Smith and colleagues built upon these and other MITF studies as a driver of intrinsic drug resistance, which is reversible and non-mutational (9). MITF and paired box 3 (PAX3) are concurrently up-regulated as an adaptive response to MAPKi and ultimately drive initial intrinsic resistance. This result was consistent with PAX3’s known function as a transcriptional regulator of MITF (10). The authors hypothesized that inhibiting MITF and PAX3 would improve MAPKi efficacy and identified nelfinavir mesylate, an HIV-1 protease inhibitor, as a potent inhibitor of those genes in a drug screen (Figure 1).

Figure 1 Improving efficacy of MAPKi with nelfinavir. MAPKi such as BRAF and MEK inhibitors lead to increased MITF and PAX3 expression. A small molecule screen against MITF and PAX3 identified nelfinavir as the most potent inhibitor. Nelfinavir in combination with MAPKi leads to increased cell death and could improve clinical response to MAPKi therapies.

Nelfinavir inhibited MITF and PAX3 expression by up-regulating the mothers against decapentaplegic homolog 2/mothers against decapentaplegic homolog 4/Ski (SMAD2/SMAD4/SKI) repressor complex. Nelfinavir also increased phosphorylated SMAD2 and SKI repressor bound to PAX3. Suppression of MITF and PAX3 by nelfinavir improved the efficacy of MAPKi by inhibiting tumor growth to a greater degree. Ectopic overexpression of MITF and PAX3 rescued the tumor’s survival ability to MAPKi. Mechanistically, mitogen-activated protein kinase kinase (MEK) suppressed PAX3 through SKI, which stimulated SMAD2 to repress the PAX3 promoter.

Nelfinavir sensitized not only BRAF- but also NRAS-mutant melanoma cells to MAPKi. Interestingly, even in melanoma cells without up-regulated MITF, the improved sensitivity to MAPKi through nelfinavir was still effective. This combination therapy is especially relevant for patients with NRAS-mutant melanomas, who have markedly worse clinical prognosis and no FDA approved targeted therapies (11). The increase in expression of MITF in NRAS-mutant melanoma cells upon MEK inhibition has been shown previously (12). Thus, Nelfinavir may also be effective in combination use with the MEK1/2 inhibitor, MEK162, to treat NRAS-mutant melanomas (13).

MITF also directly regulates peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PPARGC1α) and drives oxidative phosphorylation (14). Suppression of MITF with nelfinivir may synergize with MAPKi and inhibit aberrant oxidative metabolism, which is a significant MAPKi-acquired resistance mechanism. Altered tumor metabolism and bioenergetics are important considerations when assessing the full effects of new combinatorial therapies.

Drug repositioning, or repurposing an existing drug for a new usage, has become increasingly recognized and can provide a new source of potent inhibitors in melanoma therapy. Another example of drug repositioning is riluzole, used in treatment of amyotropic lateral sclerosis, which can inhibit cell proliferation of metabotropic glutamate receptor 1 (GRM1)-expressing melanoma cells (15). Using existing drug libraries previously unexplored for anti-tumor activity can bear new fruits of discovery.

Taken together, Smith and colleagues identify a clinically relevant combinatorial therapy through drug repositioning that could improve initial response to targeted MAPKi therapy. MITF repression has been linked to increased cell invasion and metastasis (16). Thus, there needs to be further studies to fully examine the nelfinavir and MAPKi combination. Nonetheless, this study is an important step in discovering new personalizable combinatorial treatments that could improve response to targeted therapies and perhaps even immunotherapies.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned and reviewed by the Section Editor Jie Dai (Key Laboratory of Carcinogenesis and Translational Research, Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China).

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at http://dx.doi.org/10.21037/tcr.2016.05.28). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Shi H, Hugo W, Kong X, et al. Acquired resistance and clonal evolution in melanoma during BRAF inhibitor therapy. Cancer Discov 2014;4:80-93. [Crossref] [PubMed]
  2. Zhang G, Frederick DT, Wu L, et al. Targeting mitochondrial biogenesis to overcome drug resistance to MAPK inhibitors. J Clin Invest 2016;126:1834-56. [Crossref] [PubMed]
  3. Gopal YN, Deng W, Woodman SE, et al. Basal and treatment-induced activation of AKT mediates resistance to cell death by AZD6244 (ARRY-142886) in Braf-mutant human cutaneous melanoma cells. Cancer Res 2010;70:8736-47. [Crossref] [PubMed]
  4. Sun C, Wang L, Huang S, et al. Reversible and adaptive resistance to BRAF(V600E) inhibition in melanoma. Nature 2014;508:118-22. [Crossref] [PubMed]
  5. Hu-Lieskovan S, Mok S, Homet Moreno B, et al. Improved antitumor activity of immunotherapy with BRAF and MEK inhibitors in BRAF(V600E) melanoma. Sci Transl Med 2015;7:279ra41 [Crossref] [PubMed]
  6. Lassen A, Atefi M, Robert L, et al. Effects of AKT inhibitor therapy in response and resistance to BRAF inhibition in melanoma. Mol Cancer 2014;13:83. [Crossref] [PubMed]
  7. Konieczkowski DJ, Johannessen CM, Abudayyeh O, et al. A melanoma cell state distinction influences sensitivity to MAPK pathway inhibitors. Cancer Discov 2014;4:816-27. [Crossref] [PubMed]
  8. Müller J, Krijgsman O, Tsoi J, et al. Low MITF/AXL ratio predicts early resistance to multiple targeted drugs in melanoma. Nat Commun 2014;5:5712. [Crossref] [PubMed]
  9. Smith MP, Brunton H, Rowling EJ, et al. Inhibiting Drivers of Non-mutational Drug Tolerance Is a Salvage Strategy for Targeted Melanoma Therapy. Cancer Cell 2016;29:270-84. [Crossref] [PubMed]
  10. Kubic JD, Young KP, Plummer RS, et al. Pigmentation PAX-ways: the role of Pax3 in melanogenesis, melanocyte stem cell maintenance, and disease. Pigment Cell Melanoma Res 2008;21:627-45. [Crossref] [PubMed]
  11. Jakob JA, Bassett RL Jr, Ng CS, et al. NRAS mutation status is an independent prognostic factor in metastatic melanoma. Cancer 2012;118:4014-23. [Crossref] [PubMed]
  12. Gopal YN, Rizos H, Chen G, et al. Inhibition of mTORC1/2 overcomes resistance to MAPK pathway inhibitors mediated by PGC1α and oxidative phosphorylation in melanoma. Cancer Res 2014;74:7037-47. [Crossref] [PubMed]
  13. Ascierto PA, Schadendorf D, Berking C, et al. MEK162 for patients with advanced melanoma harbouring NRAS or Val600 BRAF mutations: a non-randomised, open-label phase 2 study. Lancet Oncol 2013;14:249-56. [Crossref] [PubMed]
  14. Haq R, Shoag J, Andreu-Perez P, et al. Oncogenic BRAF regulates oxidative metabolism via PGC1α and MITF. Cancer Cell 2013;23:302-15. [Crossref] [PubMed]
  15. Namkoong J, Shin SS, Lee HJ, et al. Metabotropic glutamate receptor 1 and glutamate signaling in human melanoma. Cancer Res 2007;67:2298-305. [Crossref] [PubMed]
  16. Goodall J, Carreira S, Denat L, et al. Brn-2 represses microphthalmia-associated transcription factor expression and marks a distinct subpopulation of microphthalmia-associated transcription factor-negative melanoma cells. Cancer Res 2008;68:7788-94. [Crossref] [PubMed]
Cite this article as: Wu LW, Zhang G, Herlyn M. Repurposing an HIV drug to improve efficacy of targeted therapy in melanoma. Transl Cancer Res 2016;5(Suppl 1):S106-S108. doi: 10.21037/tcr.2016.05.28

Download Citation