v
Search
Advanced Search

Publications > Journals > Oncology Advances > Article Full Text

  • OPEN ACCESS

Expanding Horizons in Cancer Immunotherapy: The Potential of Small Molecule Immune Checkpoint Inhibitors

  • Wenxue Ma1,*  and
  • Theia Minev2
 Author information
Oncology Advances   2024;2(3):158-161

doi: 10.14218/OnA.2024.00020

The landscape of cancer treatment has witnessed a paradigm shift with the advent of immunotherapy, particularly immune checkpoint inhibitors (ICIs).1–4 Srivastava et al.’s review,5Small molecules as immune checkpoints inhibitors in cancer therapeutics”, provides an insightful and comprehensive analysis of the emerging role of small molecules in this transformative field. This commentary aimed to underscore the critical contributions of this review and discuss the broader implications for cancer therapy.

Expanding the arsenal: Small molecule ICIs

Immune checkpoint inhibitors have revolutionized cancer treatment by unleashing the body’s immune system to target and destroy cancer cells.6,7 Traditional ICIs, such as monoclonal antibodies targeting programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) and cytotoxic T-lymphocyte-associated protein 4, have been successful in treating various cancers.8,9 However, they have some limitations, including high production costs, limited tissue penetration, and significant immune-related adverse events.10,11

Srivastava et al.5 highlight small molecule ICIs as a promising alternative that addresses many of these limitations. Their review meticulously catalogs various small molecules, including those approved by the U.S. Food and Drug Administration (hereinafter referred to as FDA) and those under clinical trials, highlighting their mechanisms of action, efficacy, and potential advantages over monoclonal antibodies. Small molecules offer several benefits: they are less immunogenic, can be administered orally, and have better tissue penetration, making them an attractive option for cancer therapy.

Table 1 below summarizes the comparative advantages of small molecule ICIs over monoclonal antibodies, highlighting their potential to address many of the limitations of traditional ICI therapies.

Table 1

Comparative advantages of small molecule ICIs over monoclonal antibodies

FeatureSmall moleculesMonoclonal antibodies
AdministrationOral availabilityIntravenous infusion
Tissue penetrationBetter penetrationLimited penetration
ImmunogenicityLess immunogenicMore immunogenic
Production costLower costHigher cost
Side effectsFewer irAEsMore irAEs

Highlighting key advances

The review discusses a range of small molecules, from those disrupting PD-1/PD-L1 interactions to novel compounds targeting other immune checkpoints such as cytotoxic T-lymphocyte-associated protein 4, T cell immunoreceptor with Ig and ITIM domain, and CD47.12–16 For instance, the authors highlight FDA-approved drugs like Gefitinib and Ruxolitinib, which have shown potential in downregulating PD-L1 expression, thereby enhancing anti-tumor immunity.17,18 They also discuss emerging molecules such as INCB086550 and CA170, which are currently in clinical trials and show promise in preclinical models.8,12 An overview of the small molecules explored in this review, including their targets, clinical trial trials, and noted advantages, is presented in Table 2 below.8,17,19–41

Table 2

Summary of key small molecules discussed in the review

Small moleculeTargetClinical trial statusNoted advantagesReferences
GefitinibPD-L1FDA-approvedEnhances anti-tumor immunity by inhibiting EGF signaling and destabilizing PD-L18,17
RuxolitinibJAK-STATsFDA-approvedDownregulates PD-L1 expression in NSCLC and breast cancer cells19,20
INCB086550PD-L1Phase 2Reduces tumor growth by activating T cells and blocking PD-1/PD-L1 pathways12,21
CA170VISTA, PD-L1Phase 1 and Phase 2Dual inhibitor that upregulates PD-L1 expression shows efficacy in preclinical models and early clinical trials, orally administered Phase 1 and Phase 2 clinical trials22,23
FedratinibJAK-STATsFDA-approvedTargets PD-L1 expression, approved for myelofibrosis24,25
SelumetinibMEK1/2FDA-approvedInhibits PD-L1 in lung adenocarcinoma cells, used for neurofibromatosis type 1 in children26,27
BelinostatHDACFDA-approvedIncreases PD-L1 expression, enhances in vivo anti-PD-1/PD-L1 antibodies28,29
AzacytidineDNA hypo-methylating agentPhase 2Upregulates PD-L1 expression potentiates anti-PD-L1 antibodies in various cancer models30,31
DecitabineDNA hypo-methylating agentPhase 2Similar to Azacytidine, used in combination with PARP inhibitors for leukemia32,33
RomidepsinHDACFDA-approvedRegulates PD-L1 expression, suppresses cellular immune functions in colon cancer34,35
PanobinostatHDACFDA-approvedOral DAC inhibitor, used for multiple myeloma36,37
VorinostatHDACFDA-approvedIncreases PD-L1 expression, enhances the efficacy of anti-PD-1/PD-L1 antibodies38,39
MetforminAMPKWidely used for anti-diabetic drugActivates T-cells, prevents glycosylation of PD-L1, promotes antitumor immunity40,41

The meticulous detailing of these small molecules, supported by robust preclinical and clinical data, underscores their potential to overcome resistance mechanisms and reduce immune-related adverse events, which are significant hurdles in the current immunotherapy landscape.

Addressing challenges and future directions

While the review highlights the potential of small molecule ICIs, it also acknowledges the remaining challenges. The development of resistance, the need for biomarkers to predict response, and the management of adverse effects all require ongoing research. Srivastava et al.5 call for more in-depth clinical and biological phenotyping to optimize the therapeutic outcomes of these novel agents.11,42

The review’s comprehensive nature and forward-looking perspective make it clear that small molecule ICIs are not merely a fleeting trend but a crucial component of the future of cancer immunotherapy. The potential to combine these agents with existing therapies, including monoclonal antibodies and traditional chemotherapies, opens new avenues for synergistic effects and improved patient outcomes.

The review by Srivastava et al.5 represents a significant contribution to the field of cancer immunotherapy, offering a detailed and insightful examination of small molecule ICIs. As the field evolves, the insights presented in this review will undoubtedly serve as a foundation for future research and development. The potential of small molecule ICIs to transform cancer treatment is immense, and this review paves the way for further exploration and clinical advancements.

Declarations

Acknowledgement

None.

Funding

None.

Conflict of interest

One of the authors, Wenxue Ma has been an editorial board member of Oncology Advances since May 2021. The authors have no other conflict of interest to note.

Authors’ contributions

Conceptualization (WM), manuscript writing and editing (TM, WM). All authors have read and agreed to the published version of the manuscript.

References

  1. Shiravand Y, Khodadadi F, Kashani SMA, Hosseini-Fard SR, Hosseini S, Sadeghirad H, et al. Immune Checkpoint Inhibitors in Cancer Therapy. Curr Oncol 2022;29(5):3044-3060 View Article PubMed/NCBI
  2. Yao L, Jia G, Lu L, Bao Y, Ma W. Factors affecting tumor responders and predictive biomarkers of toxicities in cancer patients treated with immune checkpoint inhibitors. Int Immunopharmacol 2020;85:106628 View Article PubMed/NCBI
  3. Drobni ZD, Gongora C, Taron J, Suero-Abreu GA, Karady J, Gilman HK, et al. Impact of immune checkpoint inhibitors on atherosclerosis progression in patients with lung cancer. J Immunother Cancer 2023;11(7):e007307 View Article PubMed/NCBI
  4. Blum SM, Rouhani SJ, Sullivan RJ. Effects of immune-related adverse events (irAEs) and their treatment on antitumor immune responses. Immunol Rev 2023;318(1):167-178 View Article PubMed/NCBI
  5. Srivastava N, Saxena A, Saxena AK. Small Molecules as Immune Check points inhibitors in Cancer Therapeutics. Oncol Adv 2024;2(3):148-157 View Article
  6. Walsh RJ, Sundar R, Lim JSJ. Immune checkpoint inhibitor combinations-current and emerging strategies. Br J Cancer 2023;128(8):1415-1417 View Article PubMed/NCBI
  7. Baxevanis CN. Immune Checkpoint Inhibitors in Cancer Therapy-How Can We Improve Clinical Benefits?. Cancers (Basel) 2023;15(3):881 View Article PubMed/NCBI
  8. Javed SA, Najmi A, Ahsan W, Zoghebi K. Targeting PD-1/PD-L-1 immune checkpoint inhibition for cancer immunotherapy: success and challenges. Front Immunol 2024;15:1383456 View Article PubMed/NCBI
  9. Babamohamadi M, Mohammadi N, Faryadi E, Haddadi M, Merati A, Ghobadinezhad F, et al. Anti-CTLA-4 nanobody as a promising approach in cancer immunotherapy. Cell Death Dis 2024;15(1):17 View Article PubMed/NCBI
  10. Mohite P, Yadav V, Pandhare R, Maitra S, Saleh FM, Saleem RM, et al. Revolutionizing Cancer Treatment: Unleashing the Power of Viral Vaccines, Monoclonal Antibodies, and Proteolysis-Targeting Chimeras in the New Era of Immunotherapy. ACS Omega 2024;9(7):7277-7295 View Article PubMed/NCBI
  11. Yin Q, Wu L, Han L, Zheng X, Tong R, Li L, et al. Immune-related adverse events of immune checkpoint inhibitors: a review. Front Immunol 2023;14:1167975 View Article PubMed/NCBI
  12. Chen L, Zhao X, Liu X, Ouyang Y, Xu C, Shi Y. Development of small molecule drugs targeting immune checkpoints. Cancer Biol Med 2024;21(5):382-399 View Article PubMed/NCBI
  13. Chen Q, Guo X, Ma W. Opportunities and challenges of CD47-targeted therapy in cancer immunotherapy. Oncol Res 2023;32(1):49-60 View Article PubMed/NCBI
  14. Lau APY, Khavkine Binstock SS, Thu KL. CD47: The Next Frontier in Immune Checkpoint Blockade for Non-Small Cell Lung Cancer. Cancers (Basel) 2023;15(21):5229 View Article PubMed/NCBI
  15. Zhang P, Liu X, Gu Z, Jiang Z, Zhao S, Song Y, et al. Targeting TIGIT for cancer immunotherapy: recent advances and future directions. Biomark Res 2024;12(1):7 View Article PubMed/NCBI
  16. Zhao J, Li L, Yin H, Feng X, Lu Q. TIGIT: An emerging immune checkpoint target for immunotherapy in autoimmune disease and cancer. Int Immunopharmacol 2023;120:110358 View Article PubMed/NCBI
  17. Lin X, Kang K, Chen P, Zeng Z, Li G, Xiong W, et al. Regulatory mechanisms of PD-1/PD-L1 in cancers. Mol Cancer 2024;23(1):108 View Article PubMed/NCBI
  18. Yin S, Chen Z, Chen D, Yan D. Strategies targeting PD-L1 expression and associated opportunities for cancer combination therapy. Theranostics 2023;13(5):1520-1544 View Article PubMed/NCBI
  19. Xue C, Yao Q, Gu X, Shi Q, Yuan X, Chu Q, et al. Evolving cognition of the JAK-STAT signaling pathway: autoimmune disorders and cancer. Signal Transduct Target Ther 2023;8(1):204 View Article PubMed/NCBI
  20. Qureshy Z, Johnson DE, Grandis JR. Targeting the JAK/STAT pathway in solid tumors. J Cancer Metastasis Treat 2020;6:27 PubMed/NCBI
  21. Koblish HK, Wu L, Wang LS, Liu PCC, Wynn R, Rios-Doria J, et al. Characterization of INCB086550: A Potent and Novel Small-Molecule PD-L1 Inhibitor. Cancer Discov 2022;12(6):1482-1499 View Article PubMed/NCBI
  22. Sasikumar PG, Sudarshan NS, Adurthi S, Ramachandra RK, Samiulla DS, Lakshminarasimhan A, et al. PD-1 derived CA-170 is an oral immune checkpoint inhibitor that exhibits preclinical anti-tumor efficacy. Commun Biol 2021;4(1):699 View Article PubMed/NCBI
  23. Lee JJ, Powderly JD, Patel MR, Brody J, Hamilton EP, Infante JR, et al. Phase 1 trial of CA-170, a novel oral small molecule dual inhibitor of immune checkpoints PD-1 and VISTA, in patients (pts) with advanced solid tumor or lymphomas. J Clin Oncol 2017;35(15_suppl):TPS3099-TPS3099 View Article
  24. Kong T, Yu L, Laranjeira ABA, Fisher DAC, He F, Cox MJ, et al. Comprehensive profiling of clinical JAK inhibitors in myeloproliferative neoplasms. Am J Hematol 2023;98(7):1029-1042 View Article PubMed/NCBI
  25. Saha C, Harrison C. Fedratinib, the first selective JAK2 inhibitor approved for treatment of myelofibrosis - an option beyond ruxolitinib. Expert Rev Hematol 2022;15(7):583-595 View Article PubMed/NCBI
  26. Chénard-Poirier M, Hansen AR, Gutierrez ME, Rasco D, Xing Y, Chen LC, et al. A phase 1 trial of the MEK inhibitor selumetinib in combination with pembrolizumab for advanced or metastatic solid tumors. Invest New Drugs 2024;42(3):241-251 View Article PubMed/NCBI
  27. Armstrong AE, Belzberg AJ, Crawford JR, Hirbe AC, Wang ZJ. Treatment decisions and the use of MEK inhibitors for children with neurofibromatosis type 1-related plexiform neurofibromas. BMC Cancer 2023;23(1):553 View Article PubMed/NCBI
  28. Wen T, Sun G, Jiang W, He X, Shi Y, Ma F, et al. Histone deacetylases inhibitor chidamide synergizes with humanized PD1 antibody to enhance T-cell chemokine expression and augment Ifn-γ response in NK-T cell lymphoma. EBioMedicine 2023;87:104420 View Article PubMed/NCBI
  29. Shen C, Li M, Duan Y, Jiang X, Hou X, Xue F, et al. HDAC inhibitors enhance the anti-tumor effect of immunotherapies in hepatocellular carcinoma. Front Immunol 2023;14:1170207 View Article PubMed/NCBI
  30. Ebelt ND, Manuel ER. 5-Azacytidine-Mediated Modulation of the Immune Microenvironment in Murine Acute Myeloid Leukemia. Cancers (Basel) 2022;15(1):118 View Article PubMed/NCBI
  31. Wong KK, Hassan R, Yaacob NS. Hypomethylating Agents and Immunotherapy: Therapeutic Synergism in Acute Myeloid Leukemia and Myelodysplastic Syndromes. Front Oncol 2021;11:624742 View Article PubMed/NCBI
  32. Gallimore F, Fandy TE. Therapeutic Applications of Azanucleoside Analogs as DNA Demethylating Agents. Epigenomes 2023;7(3):12 View Article PubMed/NCBI
  33. Padella A, Ghelli Luserna Di Rorà A, Marconi G, Ghetti M, Martinelli G, Simonetti G. Targeting PARP proteins in acute leukemia: DNA damage response inhibition and therapeutic strategies. J Hematol Oncol 2022;15(1):10 View Article PubMed/NCBI
  34. Lian B, Chen X, Shen K. Inhibition of histone deacetylases attenuates tumor progression and improves immunotherapy in breast cancer. Front Immunol 2023;14:1164514 View Article PubMed/NCBI
  35. Shi Y, Fu Y, Zhang X, Zhao G, Yao Y, Guo Y, et al. Romidepsin (FK228) regulates the expression of the immune checkpoint ligand PD-L1 and suppresses cellular immune functions in colon cancer. Cancer Immunol Immunother 2021;70(1):61-73 View Article PubMed/NCBI
  36. Zhou YB, Zhang YM, Huang HH, Shen LJ, Han XF, Hu XB, et al. Pharmacodynamic, pharmacokinetic, and phase 1a study of bisthianostat, a novel histone deacetylase inhibitor, for the treatment of relapsed or refractory multiple myeloma. Acta Pharmacol Sin 2022;43(4):1091-1099 View Article PubMed/NCBI
  37. Eleutherakis-Papaiakovou E, Kanellias N, Kastritis E, Gavriatopoulou M, Terpos E, Dimopoulos MA. Efficacy of Panobinostat for the Treatment of Multiple Myeloma. J Oncol 2020;2020:7131802 View Article PubMed/NCBI
  38. Mei M, Chen L, Godfrey J, Song J, Egelston C, Puverel S, et al. Pembrolizumab plus vorinostat induces responses in patients with Hodgkin lymphoma refractory to prior PD-1 blockade. Blood 2023;142(16):1359-1370 View Article PubMed/NCBI
  39. Laengle J, Kabiljo J, Hunter L, Homola J, Prodinger S, Egger G, et al. Histone deacetylase inhibitors valproic acid and vorinostat enhance trastuzumab-mediated antibody-dependent cell-mediated phagocytosis. J Immunother Cancer 2020;8(1):e000195 View Article PubMed/NCBI
  40. Kim K, Yang WH, Jung YS, Cha JH. A new aspect of an old friend: the beneficial effect of metformin on anti-tumor immunity. BMB Rep 2020;53(10):512-520 View Article PubMed/NCBI
  41. Lord SR, Harris AL. Is it still worth pursuing the repurposing of metformin as a cancer therapeutic?. Br J Cancer 2023;128(6):958-966 View Article PubMed/NCBI
  42. Les I, Martínez M, Pérez-Francisco I, Cabero M, Teijeira L, Arrazubi V, et al. Predictive Biomarkers for Checkpoint Inhibitor Immune-Related Adverse Events. Cancers (Basel) 2023;15(5):1629 View Article PubMed/NCBI
  • Oncology Advances
  • eISSN 2996-3427
Back to Top

Expanding Horizons in Cancer Immunotherapy: The Potential of Small Molecule Immune Checkpoint Inhibitors

Wenxue Ma, Theia Minev
  • Reset Zoom
  • Download TIFF