SINGLE-TARGET INHIBITION OF THE PI3K/AKT/mTOR (PAM) PATHWAY LEAVES OPENINGS FOR HR+/HER2- ADVANCED/METASTATIC BREAST CANCER TO BREAK THROUGH1-4

Comprehensive inhibition of the PAM pathway is needed to overcome the limitations of current single-target inhibitors2,3,5,6

The PI3K/AKT/mTOR (PAM) pathway is a cancer control center

The PAM pathway is a driver of HR+/HER2- advanced/metastatic breast cancer, contributing to key cellular processes involved in oncogenesis and tumor progression3,7-13

Cell metabolism
Cell survival
Cell proliferation

PAM pathway hyperactivation can be an oncogenic process independent of PIK3CA mutational status.3,11,14

PI3K/AKT/mTOR (PAM) pathway crosstalk allows for adaptive resistance that maintains pathway viability1,2,4,6,11,12,15,16

The multiple components of the PAM pathway provide functional redundancy via feedback loops that limit the activity of single-target PAM pathway inhibitors.1,2,4,6

Single-target inhibition
Inhibition arrow
Adaptive resistance
PI3K-α
inhibition
  • PI3K-β signaling enhanced17-19
  • mTORC1 signaling enhanced20-22
AKT
inhibition
  • mTORC1 signaling enhanced23,24
  • RTKs are upregulated25
mTORC1
inhibition
  • PI3K-AKT signaling enhanced26-28
  • RTKs are upregulated27
PI3K/AKT/mTOR (PAM) pathway crosstalk allows for adaptive resistance that maintains pathway viability

Single-target PAM pathway inhibition allows tumors to develop escape mechanisms.

Multi-target, comprehensive PAM pathway inhibition is needed.2,3,5,6,30

Figure adapted from Glaviano A, et al. Mol Cancer. 2023;22(1):138; He Y, et al. Sig Transduct Target Ther. 2021; 425(6):1-17; Hoxhaj G, et al. Nat Rev Cancer. 2020;20(2):74-88; Manning BD, et al. Cell. 2017;169(3):381-405.
For patients with HR+/HER2- advanced/metastatic breast cancer who have progressed on endocrine-based therapy,

Current single-target inhibitors of the PI3K/AKT/mTOR (PAM) pathway fall short31-34

Many targeted therapies may offer:

Limited eligibility

Predominantly indicated for those with PIK3CA/AKT1 mutations, excluding most patients31-35

Modest efficacy

Provide modest mPFS benefit in the second-line setting31-34

Tolerability challenges

Associated with adverse events such as hyperglycemia, rash, and diarrhea, which often lead to dose interruptions, dose reductions, and discontinuations31-34

There is a need for a comprehensive PAM inhibitor that is efficacious, tolerable, and demonstrates activity in a broad population of patients, independent of PIK3CA mutational status.3,6,36

The PI3K/AKT/mTOR (PAM) pathway can drive treatment resistance to both ER- and CDK4/6-based treatments37-43

Interaction between estrogen, cyclin D1-CDK4/6, and PAM signaling pathways increases the complexity of therapeutic targeting in HR+/HER2- advanced/metastatic breast cancer.37,44,45

Estrogen, cyclin D1-CDK4/6, and PI3K/AKT/mTOR (PAM) signaling pathways interact in HR+/HER2- advanced/metastatic breast cancer

The PAM pathway can induce estrogen-dependent and estrogen-independent ER activity46-48

ER can directly activate PI3K, fueling PAM pathway activity49

The PAM and ER pathways can induce cyclin D1 upregulation, driving activation of the cyclin D1-CDK4/6 pathway50-52

Comprehensive inhibition of the PI3K/AKT/mTOR (PAM) pathway may present a new path forward in treatment

Blocking the PAM pathway comprehensively may overcome treatment resistance and enhance sensitivity to ER- and CDK4/6-based treatments, offering additional options for patients with HR+/HER2- advanced/metastatic breast cancer who have experienced disease progression.6,37,53-60

An image of a dam with 3 closed doors, stopping water from flowing through

Celcuity is closing in on the potential of comprehensive PAM pathway inhibition

Learn more about Celcuity

AKT=protein kinase B; AKT1=protein kinase B subunit 1; CDK4/6=cyclin-dependent kinases 4 and 6; ER=estrogen receptor; HER2-=human epidermal growth factor receptor 2 negative; HR+=hormone receptor positive; mPFS=median progression-free survival; mTOR=mammalian target of rapamycin; mTORC1=mammalian target of rapamycin complex 1; mTORC2=mammalian target of rapamycin complex 2; PI3K=phosphoinositide 3-kinase; PI3Kα=PI3K subunit alpha; PI3Kβ=PI3K subunit beta; PIK3CA=phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha; PTEN=phosphatase and tensin homolog; RTK=receptor tyrosine kinase.

REFERENCES: 1. Bergholz JS, Zhao JJ. How compensatory mechanisms and adaptive rewiring have shaped our understanding of therapeutic resistance in cancer. Cancer Res. 2021;81(24):6074-6077. 2. Wright SCE, Vasilevski N, Serra V, Rodon J, Eichhorn PJA. Mechanisms of resistance to PI3K inhibitors in cancer: adaptive responses, drug tolerance and cellular plasticity. Cancers (Basel). 2021;13(7):1538. 3. Browne IM, André F, Chandarlapaty S, Carey LA, Turner NC. Optimal targeting of PI3K-AKT and mTOR in advanced oestrogen receptor-positive breast cancer. Lancet Oncol. 2024;25(4):e139-e151. 4. Rozengurt E, Soares HP, Sinnet-Smith J. Suppression of feedback loops mediated by PI3K/mTOR induces multiple overactivation of compensatory pathways: an unintended consequence leading to drug resistance. Mol Cancer Ther. 2014;13(11):2477-2488. 5. Rossetti S, Broege A, Sen A, et al. Gedatolisib shows superior potency and efficacy versus single-node PI3K/AKT/mTOR inhibitors in breast cancer models. NPJ Breast Cancer. 2024;10(1):40. 6. Browne IM, Okines AFC. Resistance to targeted inhibitors of the PI3K/AKT/mTOR pathway in advanced oestrogen-receptor-positive breast cancer. Cancers (Basel). 2024;16(12):2259. 7. Liu L, Graff SL, Wang Y. New emerging therapies targeting PI3K/AKT/mTOR/PTEN pathway in hormonal receptor-positive and HER2-negative breast cancer—current state and molecular pathology perspective. Cancers (Basel). 2024;17(1):16. 8. Tortorella E, Giantulli S, Sciarra A, Silvestri I. AR and PI3K/AKT in prostate cancer: a tale of two interconnected pathways. Int J Mol Sci. 2023;24(3):2046. 9. Paplomata E, O’Regan R. The PI3K/AKT/mTOR pathway in breast cancer: targets, trials and biomarkers. Ther Adv Med Oncol. 2014;6(4):154-166. 10. Vasan N, Cantley LC. At a crossroads: how to translate the roles of PI3K in oncogenic and metabolic signalling into improvements in cancer therapy. Nat Rev Clin Oncol. 2022;19(7):471-485. 11. Glaviano A, Foo ASC, Lam HY, et al. PI3K/AKT/mTOR signaling transduction pathway and targeted therapies in cancer. Mol Cancer. 2023;22(1):138. 12. He Y, Sun MM, Zhang GG, et al. Targeting PI3K/Akt signal transduction for cancer therapy. Signal Transduct Target Ther. 2021;6(1):425. 13. du Rusquec P, Blonz C, Frenel JS, Campone M. Targeting the PI3K/Akt/mTOR pathway in estrogen-receptor positive HER2 negative advanced breast cancer. Ther Adv Med Oncol. 2020;12:1758835920940939. 14. Zhang Y, Kwok-Shing Ng P, Kucherlapati M, et al. A pan-cancer proteogenomic atlas of PI3K/AKT/mTOR pathway alterations. Cancer Cell. 2017;31(6):820-832.e3. 15. Hoxhaj G, Manning BD. The PI3K-AKT network at the interface of oncogenic signalling and cancer metabolism. Nat Rev Cancer. 2020;20(2):74-88. 16. Manning BD, Toker A. AKT/PKB signaling: navigating the network. Cell. 2017;169(3):381-405. 17. Schwartz S, Wongvipat J, Trigwell CB, et al. Feedback suppression of PI3Ka signaling in PTEN-mutated tumors is relieved by selective inhibition of PI3Kb. Cancer Cell. 2015;27(1):109-122. 18. Mao N, Zhang Z, Lee YS, et al. Defining the therapeutic selective dependencies for distinct subtypes of PI3K pathway-altered prostate cancers. Nat Commun. 2021;12(1):5053. 19. Costa C, Ebi H, Martini M, et al. Measurement of PIP3 levels reveals an unexpected role for p110β in early adaptive responses to p110α-specific inhibitors in luminal breast cancer. Cancer Cell. 2015;27(1):97-108. 20. Elkabets M, Vora S, Juric D, et al. mTORC1 inhibition is required for sensitivity to PI3K p110α inhibitors in PIK3CA-mutant breast cancer. Sci Transl Med. 2013;5(196):196ra99. 21. Gremke N, Besong I, Stroh A, et al. Targeting PI3K inhibitor resistance in breast cancer with metabolic drugs. Signal Transduct Target Ther. 2025;10(1):92. 22. Castel P, Ellis H, Bago R, et al. PDK1-SGK1 signaling sustains AKT-independent mTORC1 activation and confers resistance to PI3Kα inhibition. Cancer Cell. 2016;30(2):229-242. 23. Sobsey CA, Froehlich BC, Mitsa G, et al. mTORC1-driven protein translation correlates with clinical benefit of capivasertib within a genetically preselected cohort of PIK3CA-altered tumors. Cancer Res Commun. 2024;4(8):2058-2074. 24. Bago R, Sommer E, Castel P, et al. The hVps34-SGK3 pathway alleviates sustained PI3K/Akt inhibition by stimulating mTORC1 and tumour growth. EMBO J. 2016;35(17):1902-1922. 25. Chandarlapaty S, Sawai A, Scaltriti M, et al. AKT inhibition relieves feedback suppression of receptor tyrosine kinase expression and activity. Cancer Cell. 2011;19(1):58-71. 26. Mukherjee R, Vanaja KG, Boyer JA, et al. Regulation of PTEN translation by PI3K signaling maintains pathway homeostasis. Mol Cell. 2021;81(4):708-723.e5. 27. O’Reilly KE, Rojo F, She QB, et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 2006;66(3):1500-1508. 28. Rodrik-Outmezguine VS, Chandarlapaty S, Pagano NC, et al. mTOR kinase inhibition causes feedback-dependent biphasic regulation of AKT signaling. Cancer Discov. 2011;1(3):248-259. 29. Thorpe LM, Yuzugullu H, Zhao JJ. PI3K in cancer: divergent roles of isoforms, modes of activation and therapeutic targeting. Nat Rev Cancer. 2015;15(1):7-24. 30. Tyrakis PA, Kampjut D, Steele GF, et al. Multi-node inhibition targeting mTORC1, mTORC2 and PI3Kα potently inhibits the PI3K/AKT/mTOR pathway in endometrial and breast cancer models. Br J Cancer. 2025;133(2):144-154. 31. Piqray (alpelisib) [package insert]. East Hanover, NJ: Novartis Pharmaceuticals Corporation. 2024. 32. Itovebi (inavolisib) [package insert]. South San Francisco, CA: Genentech USA, Inc. 2025. 33. Truqap (capivasertib) [package insert]. Wilmington, DE: AstraZeneca Pharmaceuticals LP. 2025. 34. Afinitor (everolimus) [package insert]. East Hanover, NJ: Novartis Pharmaceuticals Corporation. 2025. 35. Garg P, Ramisetty S, Nair M, et al. Strategic advancements in targeting the PI3K/AKT/mTOR pathway for breast cancer therapy. Biochem Pharmacol. 2025;236:116850. 36. Castel P, Toska E, Engelman JA, Scaltriti M. The present and future of PI3K inhibitors for cancer therapy. Nat Cancer. 2021;2(6):587-597. 37. Alves CL, Ditzel HJ. Drugging the PI3K/AKT/mTOR pathway in ER+ breast cancer. Int J Mol Sci. 2023;24(5):4522. 38. Dong C, Wu J, Chen Y, Nie J, Chen C. Activation of PI3K/AKT/mTOR pathway causes drug resistance in breast cancer. Front Pharmacol. 2021:12:628690. 39. Gao Y, Yu Y, Zhang M, Yu W, Kang L. Mechanisms of endocrine resistance in hormone receptor-positive breast cancer. Front Oncol. 2024:14:1448687. 40. Glaviano A, Wander SA, Baird RD, et al. Mechanisms of sensitivity and resistance to CDK4/CDK6 inhibitors in hormone receptor-positive breast cancer treatment. Drug Resist Updat. 2024:76:101103. 41. Goel S, Bergholz JS, Zhao JJ. Targeting cyclin-dependent kinases 4 and 6 in cancer. Nat Rev Cancer. 2022;22(6):356-372. 42. Hanker AB, Sudhan DR, Arteaga CL. Overcoming endocrine resistance in breast cancer. Cancer Cell. 2020;37(4):496-513. 43. Marra A, Trapani D, Ferraro E, Curigliano G. Mechanisms of endocrine resistance in hormone receptor-positive breast cancer. Cancer Treat Res. 2023:188:219-235. 44. Clark AS, Makhlin I, DeMichele A. Setting the pick: can PI3K inhibitors circumvent CDK 4/6 inhibitor resistance? Clin Cancer Res. 2021;27(2):371-373. 45. Cortés J, Im SA, Holgado E, Perez-Garcia JM, Schmid P, Chavez-MacGregor M. The next era of treatment for hormone receptor-positive, HER2-negative advanced breast cancer: triplet combination-based endocrine therapies. Cancer Treat Rev. 2017;61:53-60. 46. Alayev A, Salamon RS, Berger SM, et al. mTORC1 directly phosphorylates and activates ERα upon estrogen stimulation. Oncogene. 2016;35(27):3535-3543. 47. Yamnik RL, Digilova A, Davis DC, Brodt ZN, Murphy CJ, Holz MK. S6 kinase 1 regulates estrogen receptor alpha in control of breast cancer cell proliferation. J Biol Chem. 2009;284(10):6361-6369. 48. Campbell RA, Bhat-Nakshatri P, Patel NM, Constantinidou D, Ali S, Nakshatri H. Phosphatidylinositol 3-kinase/AKT-mediated activation of estrogen receptor alpha: a new model for anti-estrogen resistance. J Biol Chem. 2001;276(13):9817-9824. 49. Castoria G, Migliaccio A, Bilancio A, et al. PI3-kinase in concert with Src promotes the S-phase entry of oestradiol-stimulated MCF-7 cells. EMBO J. 2001;20(21):6050-6059. 50. Averous J, Fonseca BD, Proud CG. Regulation of cyclin D1 expression by mTORC1 signaling requires eukaryotic initiation factor 4E-binding protein 1. Oncogene. 2008;27(8):1106-13. 51. Prall OW, Sarcevic B, Musgrove EA, Watts CK, Sutherland RL. Estrogen-induced activation of Cdk4 and Cdk2 during G1-S phase progression is accompanied by increased cyclin D1 expression and decreased cyclin-dependent kinase inhibitor association with cyclin E-Cdk2. J Biol Chem. 1997;272(16):10882-10894. 52. Sabbah M, Courilleau D, Mester J, Redeuilh G. Estrogen induction of the cyclin D1 promoter: involvement of a cAMP response-like element. Proc Natl Acad Sci USA. 1999;96(20):11217–11222. 53. Broege A, Rossetti S, Sen A, et al. Functional analysis of the PI3K/AKT/mTOR pathway inhibitor, gedatolisib, plus fulvestrant with and without palbociclib in breast cancer models. Int J Mol Sci. 2025;26(12):5844. 54. Ribas R, Pancholi S, Guest SK, et al. AKT antagonist AZD5363 influences estrogen receptor function in endocrine-resistant breast cancer and synergizes with fulvestrant (ICI182780) in vivo. Mol Cancer Ther. 2015;14(9):2035-2048. 55. Alves CL, Ehmsen S, Terp MG, et al. Co-targeting CDK4/6 and AKT with endocrine therapy prevents progression in CDK4/6 inhibitor and endocrine therapy-resistant breast cancer. Nat Commun. 2021;12(1):5112. 56. Cai Z, Wang J, Li Y, et al. Overexpressed cyclin D1 and CDK4 proteins are responsible for the resistance to CDK4/6 inhibitor in breast cancer that can be reversed by PI3K/mTOR inhibitors. Sci China Life Sci. 2023;66(1):94-109. 57. Costa C, Wang Y, Ly A, et al. PTEN loss mediates clinical cross-resistance to CDK4/6 and PI3Kα inhibitors in breast cancer. Cancer Discov. 2020;10(1):72-85. 58. Hopcroft L, Wigmore EM, Williamson SC, et al. Combining the AKT inhibitor capivasertib and SERD fulvestrant is effective in palbociclib-resistant ER+ breast cancer preclinical models. NPJ Breast Cancer. 2023;9(1):64. 59. Jansen VM, Bhola NE, Bauer JA, et al. Kinome-wide RNA interference screen reveals a role for PDK1 in acquired resistance to CDK4/6 inhibition in ER-positive breast cancer. Cancer Res. 2017;77(9):2488-2499. 60. Karimi L, Alves CL, Terp MG, et al. Triple combination targeting PI3K, ER, and CDK4/6 inhibits growth of ER-positive breast cancer resistant to fulvestrant and CDK4/6 or PI3K inhibitor. Cancer Commun (Lond). 2023;43(6):720-725.