Search

Recent Articles

IJMPO—A Journey of a Thousand Miles

Author : Padmaj S. Kulkarni

Coronavirus Disease 2019 Treatment—T-Cells Hold the Key in Severe Cases

Author : Kunal Das, Nitika Agrawal, Mansi Kala, Rakhee Khanduri

Why Is China Importing COVID-19 Vaccine Now?

Author : Purvish M. Parikh

Selective cyclin-dependent kinase 4/6 inhibitors as anticancer drugs: Moving beyond hormone receptor-positive breast cancer

CC BY-NC-ND 4.0 · Indian J Med Paediatr Oncol 2019; 40(01): 15-20

DOI: DOI: 10.4103/ijmpo.ijmpo_87_18

Abstract

The cyclin D-cyclin-dependent kinase (CDK) 4/6 pathway controls the cell cycle machinery by regulating the G1-to-S-phase transition. Dysregulation of this pathway, resulting in increased cellular proliferation, is frequently observed in a variety of human cancers. Activation of cyclin D-CDK 4/6 pathway can occur through different mechanisms, including gene amplification/rearrangement, loss of negative regulatory factors, epigenetic modifications, and point mutations of different components of this pathway. Quite conspicuously, CDK 4/6 inhibitors have emerged as promising anticancer agents in various tumors in which CDK 4/6 has a pivotal role in the G1-to-S-phase cell cycle transition. The clinical use of first-generation, nonselective pan-CDK inhibitors was not progressed beyond early phase trials, due to unacceptable toxicity and lack of efficacy noted with these agents. The emergence of selective CDK 4/6 inhibitors, including ribociclib, abemaciclib, and palbociclib, has enabled us to effectively target cyclin D-CDK 4/6 pathway, at the cost of acceptable toxicity. The results of landmark phase III trials investigating palbociclib and ribociclib in advanced hormone receptor (HR)-positive breast cancer have demonstrated a substantial clinical benefit with a well-tolerated toxicity profile. Mechanisms of acquired resistance to selective CDK 4/6 inhibitors are beginning to emerge. Clearly, a detailed understanding of these resistance mechanisms is very much essential for the rational development of post-CDK 4/6 inhibitor therapeutic strategies. Extending the use of selective CDK 4/6 inhibitors beyond HR-positive breast cancer is a challenging task and will likely require identification of clinically meaningful biomarkers to predict response and the use of combination approaches to optimize CDK 4/6 targeting.



Publication History

Article published online:
08 June 2021

© 2019. Indian Society of Medical and paediatric Oncology. This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial-License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/).

Thieme Medical and Scientific publishers pvt. Ltd.
A-12, 2nd Floor, Sector 2, Noida-201301 Up, India

Abstract

The cyclin D-cyclin-dependent kinase (CDK) 4/6 pathway controls the cell cycle machinery by regulating the G1-to-S-phase transition. Dysregulation of this pathway, resulting in increased cellular proliferation, is frequently observed in a variety of human cancers. Activation of cyclin D-CDK 4/6 pathway can occur through different mechanisms, including gene amplification/rearrangement, loss of negative regulatory factors, epigenetic modifications, and point mutations of different components of this pathway. Quite conspicuously, CDK 4/6 inhibitors have emerged as promising anticancer agents in various tumors in which CDK 4/6 has a pivotal role in the G1-to-S-phase cell cycle transition. The clinical use of first-generation, nonselective pan-CDK inhibitors was not progressed beyond early phase trials, due to unacceptable toxicity and lack of efficacy noted with these agents. The emergence of selective CDK 4/6 inhibitors, including ribociclib, abemaciclib, and palbociclib, has enabled us to effectively target cyclin D-CDK 4/6 pathway, at the cost of acceptable toxicity. The results of landmark phase III trials investigating palbociclib and ribociclib in advanced hormone receptor (HR)-positive breast cancer have demonstrated a substantial clinical benefit with a well-tolerated toxicity profile. Mechanisms of acquired resistance to selective CDK 4/6 inhibitors are beginning to emerge. Clearly, a detailed understanding of these resistance mechanisms is very much essential for the rational development of post-CDK 4/6 inhibitor therapeutic strategies. Extending the use of selective CDK 4/6 inhibitors beyond HR-positive breast cancer is a challenging task and will likely require identification of clinically meaningful biomarkers to predict response and the use of combination approaches to optimize CDK 4/6 targeting.


Introduction

Uncontrolled cellular proliferation, as a result of dysregulated cell division, is one of the key hallmarks of cancer, and identifying appropriate therapeutic targets to block cell division is a widely used strategy of anticancer therapy. Cyclin-dependent kinases (CDKs) control the transition from one stage of the cell cycle to the next, and they are activated upon interaction with their partner cyclins.[1] Therefore, quite conspicuously, CDKs have long been regarded as attractive therapeutic targets for cancer treatment. Unfortunately, many of the early first-generation CDK inhibitors failed in the clinical development because of nonselective pan-CDK inhibition, which was found to be toxic-to-nonmalignant cells.[2] These issues of effectiveness and toxicity of nonselective CDK inhibitors seem to have been overcome in the last decade by the development of selective CDK-targeting agents – which selectively target CDK 4/6.

Dysregulation of cyclin D-CDK 4/6 pathway is frequently observed in human cancers and results in uncontrolled cell cycle progression.[3] CDK 4/6 mediates the transition from G1 to S phase by associating with cyclin-D and regulating the phosphorylation of retinoblastoma (Rb) protein. Increased cyclin D-CDK 4/6 pathway activity can occur through several mechanisms, including overexpression of D-type cyclins, mutation or amplification of CDK 4/6, epigenetic alterations, or loss of negative regulators.[2],[3] Thus, the development of selective CDK 4/6 inhibitors offers a novel therapeutic approach in the field of oncology. Following the encouraging results of early phase clinical trials, three of the selective CDK 4/6 inhibitors (e.g., abemaciclib, palbociclib, and ribociclib) have emerged as agents with promising anticancer activity and acceptable toxicity profile,[4],[5],[6],[7],[8],[9],[10] and among them, palbociclib and ribociclib have already received FDA approval, with landmark phase III data available, in the setting of hormone receptor (HR)-positive, human epidermal growth factor receptor-2 (HER-2)-negative advanced breast cancer.[11],[12],[13],[14]

In this review, we discuss the rationale of selectively targeting CDK 4/6 pathway and the challenges with regard to optimizing their use. We also provide an overview of the currently available clinical data for selective CDK4/6 inhibitors in different human cancers, other than HR-positive, HER-2 negative breast cancer.


Overview of Cyclin D-Cyclin-Dependent Kinase 4/6 pathway Dysregulation

principle mechanisms by which the cyclin D-CDK 4/6 pathway can become dysregulated in various human cancers are amplification of the genes encoding cyclin D1 (CCND1) or deletion of the locus encoding CDKN2A. According to the published data, amplification of CCND1 is frequently found in some human cancers, for example, breast cancer (35% of cases), head-and-neck cancer (26%–39%), endometrial cancer (26%), pancreatic adenocarcinoma (25%), and nonsmall cell lung cancer (NSCLC) (5%–30%).[15],[16] In a recently reported landmark study, which investigated the role of routine molecular screening to identify actionable mutations in advanced refractory cancer patients, Cassier et al. found CCND1 amplification and homozygous deletion of CDKN2A in 17% and 21% of patients, respectively.[17]

The cyclin D-CDK 4/6 pathway can be dysregulated by multiple other mechanisms also, for example, mutations in the genes encoding various components of this pathway, epigenetic alterations, and mutations in the upstream factors. Haluska and Hodi found that about 20% of familial malignant melanoma cases harbor CDKN2A mutations.[18],[19] Epigenetic modifications of the CDKN2A gene have been reported in human ovarian cancer.[20] Jackson et al. highlighted the importance of mutations in the upstream factors as a mechanism of cyclin D-CDK 4/6 pathway dysregulation in malignant rhabdoid tumors, where the INI1/SMARCB1 gene is frequently mutated.[21]


Biologic Rationale of Selectively Inhibiting Cyclin-Dependent Kinase 4/6 in Human Cancers

The ideal CDK-targeted agents should block CDK-mediated signaling in malignant cells and at the same time should spare the aspects of CDK activity which are critical for the survival of nonmalignant cells, thus avoiding toxicity. Inhibition of CDK1 by nonspecific inhibitors could affect all cell types and result in toxicity, as evidenced by the reported fact that mouse embryos lacking CDK1 fail to develop beyond the blastocyst stage.[22] In addition, nonspecific targeting of CDKs might also result in inhibition of CDKs 7, 8, and 9, the exact functions of which are less well established.[23] Clearly, toxicity is a major concern regarding nonselective CDK-targeted agents because CDKs play a critical role in the proliferation of both normal cells and cancer cells.

The difficulty in finding a therapeutic window wherein CDK inhibition is both safe and effective was reflected in the early clinical experience with various nonselective CDK inhibitors, for example, flavopiridol and seliciclib. To date, the most well-studied nonselective CDK inhibitor is flavopiridol, which showed limited clinical benefit, mainly because of its complex pharmacokinetics and high levels of off-target effects.[24] Seliciclib, a purine-based compound that inhibits CDKs 1, 2, 5, 7, and 9, failed to demonstrate effective clinical activity in phase I studies.[25]

It is possible that cancers with known aberrations in the cyclin D-CDK 4/6 pathway will be more sensitive to CDK 4/6 inhibition than normal cells.[26] Furthermore, selective inhibitors spare CDK2 activity which allows normal cells to continue to function and proliferate. In addition, in contrast to the cytotoxic effects of pan-CDK inhibitors, selective CDK 4/6 inhibitors are usually found to have cytostatic effects, which might further limit the potential of these agents to cause significant clinical toxicity.[27]


Selective Cyclin-Dependent Kinase 4/6 Inhibitors in Cancer Therapy

As discussed earlier, after the encouraging results from preclinical studies, three CDK4/6 inhibitors have currently reached early phase clinical trials – abemaciclib, palbociclib, and ribociclib with published phase III data available for palbociclib and ribociclib, in the setting of HR-positive, HER-2-negative advanced breast cancer.[11],[12],[13],[14]

The next part of this review will focus on the currently available preclinical and clinical data of selective CDK 4/6 inhibitors in different human cancers, other than the archetypal model of ER-positive, HER-2-negative luminal breast cancer.


preclinical Data

Abemaciclib

It has been shown to reduce the phosphorylation of Rb1 in colorectal cancer and melanoma xenografts, thus inducing G1 arrest.[28] Abemaciclib has also been demonstrated to induce growth regression in vemurafenib-resistant melanoma models, in which expression of cyclin D1 was noted to be elevated in conjunction with mitogen-activated protein kinase (MApK) pathway reactivation in vitro.[29]


Ribociclib

Single-agent ribociclib has been shown to inhibit the growth of neuroblastoma and liposarcoma cell lines, by inducing G1 arrest and reducing Rb1 phosphorylation.[30] It inhibits CDK 4/6 effectively even at nanomolar concentrations.


palbociclib

It has been shown to be active in mantle cell lymphoma xenografts[31] and glioblastoma cell lines.[32] Moreover, activity of palbociclib in combination with bortezomib has been demonstrated in both acute myeloid leukemia and myeloma.[33],[34] In ovarian cancer cell lines, a response to palbociclib was found to be most marked in Rb1-proficient cell lines with low p16INK4A expression, and amplification of cyclin E1 was associated with resistance.[35]


Data from Early phase Clinical Trials

After the publication of promising results from preclinical research, quite conspicuously, selective CDK 4/6 inhibitors have been investigated in early phase clinical trials also.

Abemaciclib

The first-in-human phase I trial of abemaciclib enrolled 75 patients with advanced solid tumors.[4] The dose-limiting toxicity was Grade 3 fatigue. The most common treatment-related adverse events (AEs) included diarrhea (52%), nausea (32%), fatigue (21%), vomiting (21%), and neutropenia (19%). pharmacodynamic evidence of targeted CDK4/6 inhibition was observed, as shown by a decrease in Rb phosphorylation in the skin. In an expansion cohort of this trial in patients with NSCLC, 51

  • References

  1. Hartwell LH, Culotti J, pringle JR, Reid BJ. Genetic control of the cell division cycle in yeast. Science 1974; 183: 46-51
  2. Malumbres M, Barbacid M. Cell cycle, CDKs and cancer: A changing paradigm. Nat Rev Cancer 2009; 9: 153-66
  3. Shapiro GI. Cyclin-dependent kinase pathways as targets for cancer treatment. J Clin Oncol 2006; 24: 1770-83
  4. Shapiro G, Rosen LS, Tolcher AW, Goldman JW, Gandhi L, papadopoulos Kp. A first-in-human phase I study of the CDK4/6 inhibitor, LY2835219, for patients with advanced cancer. J Clin Oncol. 2013:31.
  5. Goldman JW, Gandhi L, patnaik A, Rosen LS, Hilton JF, papadopoulos Kp. et al. Clinical activity of LY2835219, a novel cell cycle inhibitor selective for CDK4 and CDK6, in patients with non-small cell lung cancer. J Clin Oncol. 2014:32.
  6. Infante JR, Shapiro G, Witteveen p, Gerecitano JF, Ribrag V, Chugh R. et al. A phase I study of the single-agent CDK4/6 inhibitor LEE011 in pts with advanced solid tumors and lymphomas. J Clin Oncol. 2014:32
  7. Sosman JA, Kittaneh M, Lolkema Mp, postow MA, Schwartz G, Franklin C. et al. A phase 1b/2 study of LEE011 in combination with binimetinib (MEK162) in patients with NRAS-mutant melanoma: Early encouraging clinical activity. J Clin Oncol. 2014:32.
  8. Flaherty KT, Lorusso pM, Demichele A, Abramson VG, Courtney R, Randolph SS. et alphase I, dose-escalation trial of the oral cyclin-dependent kinase 4/6 inhibitor pD 0332991, administered using a 21-day schedule in patients with advanced cancer. Clin Cancer Res 2012; 18: 568-76
  9. Schwartz GK, LoRusso pM, Dickson MA, Randolph SS, Shaik MN, Wilner KD. et alphase I study of pD 0332991, a cyclin-dependent kinase inhibitor, administered in 3-week cycles (Schedule 2/1). Br J Cancer 2011; 104: 1862-8
  10. Leonard Jp, LaCasce AS, Smith MR, Noy A, Chirieac LR, Rodig SJ. et alSelective CDK4/6 inhibition with tumor responses by pD0332991 in patients with mantle cell lymphoma. Blood 2013; 119: 4597-607
  11. Finn RS, Martin M, Rugo HS, Jones SE, Seock-Ah IM, Karen A. et al. pALOMA2: primary results from a phase III trial of palbociclib (p) with letrozole (L) compared with letrozole alone in postmenopausal women with ER+/HER2 – Advanced breast cancer (ABC). J Clin Oncol. 2016:34.
  12. Turner NC, Ro J, André F, Loi S, Verma S, Iwata H. et alpalbociclib in hormone-receptor-positive advanced breast cancer. N Engl J Med 2015; 373: 209-19
  13. Cristofanilli M, Turner NC, Bondarenko I, Ro J, Im SA, Masuda N. et alFulvestrant plus palbociclib versus fulvestrant plus placebo for treatment of hormone-receptor-positive, HER2-negative metastatic breast cancer that progressed on previous endocrine therapy (pALOMA-3): Final analysis of the multicentre, double-blind, phase 3 randomised controlled trial. Lancet Oncol 2016; 17: 425-39
  14. Hortobagyi GN, Stemmer SM, Burris HA, Yap YS, Sonke GS, Shimon Sp. et alRibociclib as first-line therapy for Hr-positive, advanced breast cancer. N Eng J Med 2017; 376: 288-9
  15. Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature 2012; 490: 61-70
  16. Musgrove EA, Caldon CE, Barraclough J, Stone A, Sutherland RL. Cyclin D as a therapeutic target in cancer. Nat Rev Cancer 2011; 11: 558-72
  17. Cassier p, Trédan O, Seigne C, Lavergne E, Fayette J, Desseigne F. et al. Identifying actionable targets in advanced cancer patients: preliminary results from the profiler program. J Clin Oncol. 2014:32.
  18. Haluska FG, Hodi FS. Molecular genetics of familial cutaneous melanoma. J Clin Oncol 1998; 16: 670-82
  19. Cairns p, polascik TJ, Eby Y, Tokino K, Califano J, Merlo A. et alFrequency of homozygous deletion at p16/CDKN2 in primary human tumours. Nat Genet 1995; 11: 210-2
  20. Abou-Zeid AA, Azzam AZ, Kamel NA. Methylation status of the gene promoter of cyclin-dependent kinase inhibitor 2A (CDKN2A) in ovarian cancer. Scand J Clin Lab Invest 2011; 71: 542-7
  21. Jackson EM, Sievert AJ, Gai X, Hakonarson H, Judkins AR, Tooke L. et alGenomic analysis using high-density single nucleotide polymorphism-based oligonucleotide arrays and multiplex ligation-dependent probe amplification provides a comprehensive analysis of INI1/SMARCB1 in malignant rhabdoid tumors. Clin Cancer Res 2009; 15: 1923-30
  22. Santamaría D, Barrière C, Cerqueira A, Hunt S, Tardy C, Newton K. et alCdk1 is sufficient to drive the mammalian cell cycle. Nature 2007; 448: 811-5
  23. Wallenfang MR, Seydoux G. Cdk-7 is required for mRNA transcription and cell cycle progression in Caenorhabditis elegans embryos. proc Natl Acad Sci U S A 2002; 99: 5527-32
  24. Dickson MA. Molecular pathways: CDK4 inhibitors for cancer therapy. Clin Cancer Res 2014; 20: 3379-83
  25. Benson C, White J, De Bono J, O'Donnell A, Raynaud F, Cruickshank C. et alA phase I trial of the selective oral cyclin-dependent kinase inhibitor seliciclib (CYC202; R-Roscovitine), administered twice daily for 7 days every 21 days. Br J Cancer 2007; 96: 29-37
  26. Roberts RJ, Bisi JE, Strum JC, Combest AJ, Darr DB, Usary JE. et alMultiple roles of cyclin-dependent kinase 4/6 inhibitors in cancer therapy. J Natl Cancer Inst 2012; 104: 476-87
  27. Choi YJ, Li X, Hydbring p, Sanda T, Stefano J, Christie AL. et alThe requirement for cyclin D function in tumor maintenance. Cancer Cell 2012; 22: 438-51
  28. Tate SC, Cai S, Ajamie RT, Burke T, Beckmann Rp, Chan EM. et alSemi-mechanistic pharmacokinetic/pharmacodynamic modeling of the antitumor activity of LY2835219, a new cyclin-dependent kinase 4/6 inhibitor, in mice bearing human tumor xenografts. Clin Cancer Res 2014; 20: 3763-74
  29. Yadav V, Burke TF, Huber L, DeshVan Hornpande RD, Zhang Y, Buchanan SG. et alThe CDK4/6 inhibitor LY2835219 overcomes vemurafenib resistance resulting from MApK reactivation and cyclin D1 upregulation. Mol Cancer Ther 2014; 13: 2253-63
  30. Rader J, Russell MR, Hart LS, Nakazawa MS, Belcastro TM, Martinez D. et alDual CDK4/CDK6 inhibition induces cell-cycle arrest and senescence in neuroblastoma. Clin Cancer Res 2013; 19: 6173-82
  31. Marzec M, Kasprzycka M, Lai R, Gladden AB, Wlodarski p, Tomczak E. et alMantle cell lymphoma cells express predominantly cyclin D1a isoform and are highly sensitive to selective inhibition of CDK4 kinase activity. Blood 2006; 108: 1744-50
  32. Wiedemeyer WR, Dunn IF, Quayle SN, Zhang J, Chheda MG, Dunn Gp. et alpattern of retinoblastoma pathway inactivation dictates response to CDK4/6 inhibition in GBM. proc Natl Acad Sci U S A 2010; 107: 11501-6
  33. Menu E, Garcia J, Huang X, Di Liberto M, Toogood pL, Chen I. et alA novel therapeutic combination using pD 0332991 and bortezomib: Study in the 5T33MM myeloma model. Cancer Res 2008; 68: 5519-23
  34. Wang L, Wang J, Blaser BW, Duchemin AM, Kusewitt DF, Liu T. et alpharmacologic inhibition of CDK4/6: Mechanistic evidence for selective activity or acquired resistance in acute myeloid leukemia. Blood 2007; 110: 2075-83
  35. Konecny GE, Winterhoff B, Kolarova T, Qi J, Manivong K, Dering J. et alExpression of p16 and retinoblastoma determines response to CDK4/6 inhibition in ovarian cancer. Clin Cancer Res 2011; 17: 1591-602
  36. Geoerger B, Bourdeaut F, DuBois SG, DeWire MD, Marabelle A, pearson AD. et alphase I study of LEE011 (CDK4/6 inhibitor) in patients with malignant rhabdoid tumors, neuroblastoma, and cyclin D-CDK4/6 pathway-activated tumors. Ann Oncol 2014; 25 Suppl 4: 455
  37. Vaughn DJ, Flaherty K, Lal p, Gallagher M, O'Dwyer p, Wilner K. et alTreatment of growing teratoma syndrome. N Engl J Med 2009; 360: 423-4
  38. Vaughn DJ, Hwang WT, Lal p, Rosen MA, Gallagher M, O'Dwyer pJ. et alphase 2 trial of the cyclin-dependent kinase 4/6 inhibitor palbociclib in patients with retinoblastoma protein-expressing germ cell tumors. Cancer 2015; 121: 1463-8
  39. Dickson MA, Tap WA, Keohan ML, D'Angelo Sp, Gounder MM, Antonescu CR. et alphase II trial of the CDK4 inhibitor pD0332991 in patients with advanced CDK4-amplified well-differentiated or dedifferentiated liposarcoma. J Clin Oncol 2013; 31: 2024-8
  40. Gopalan pK, pinder MC, Chiappori A, Ivey AM, Villegas AG, Kaye FJ. A phase II clinical trial of the CDK 4/6 inhibitor palbociclib (pD 0332991) in previously treated, advanced non-small cell lung cancer (NSCLC) patients with inactivated CDKN2A. J Clin Oncol. 2014:32
  41. Niesvizky R, Badros AZ, Costa LJ, Ely SA, Singhal SB, Stadtmauer EA. et alphase 1/2 study of cyclin-dependent kinase (CDK) 4/6 inhibitor palbociclib (pD-0332991) with bortezomib and dexamethasone in relapsed/refractory multiple myeloma. Leuk Lymphoma 2015; 56: 3320-8
  42. Chiron D, Di Liberto M, Martin p, Huang X, Sharman J, Blecua p. et alCell-cycle reprogramming for pI3K inhibition overrides a relapse-specific C481S BTK mutation revealed by longitudinal functional genomics in mantle cell lymphoma. Cancer Discov 2014; 4: 1022-35
  43. Kim S, Loo S, Chopra R, Caponigro G, Huang A, Vora S. et al. LEE011: An orally bioavailable, selective small molecule inhibitor of CDK4/6 – Reactivating Rb in cancer. Mol Cancer Ther. 2013:12.
  44. Taylor M, Sosman J, Gonzalez R, Carlino M, Kittaneh M, Lolkema M. et al. phase Ib/II study of LEE011 (CDK4/6 inhibitor) and LGX818 (BRAF inhibitor) in BRAF-mutant melanoma. Ann Oncol. 2014:25.
  45. Abukhdeir AM, Vitolo MI, Argani p, De Marzo AM, Karakas B, Konishi H. et alTamoxifen-stimulated growth of breast cancer due to p21 loss. proc Natl Acad Sci U S A 2008; 105: 288-93