Are Cyclin D and cdk Inhibitors A Good Target for Chemotherapy?
Curator: Stephen J. Williams, Ph.D.
UPDATED 7/12/2022
see below for great review
CDK4 and CDK6 kinases: From basic science to cancer therapy
Targeting cyclin-dependent kinases
Structured Abstract
BACKGROUND
ADVANCES
OUTLOOK

Abstract

Cyclin D–CDK4/6 in cancer
CDK4/6 functions in cell proliferation and oncogenesis
Mechanism of action of CDK4/6 inhibitors
Name of compound | IC50 | Other known targets | Stage of clinical development |
---|---|---|---|
Palbociclib (PD-0332991) | D1-CDK4, 11 nM; D2-CDK6, 15 nM; D3-CDK4, 9 nM |
FDA-approved for HR+/HER2– advanced breast cancer in combination with endocrine therapy; phase 2/3 trials for several other tumor types |
|
Ribociclib (LEE011) | D1-CDK4, 10 nM; D3-CDK6, 39 nM |
FDA-approved for HR+/HER2– advanced breast cancer in combination with endocrine therapy; phase 2/3 trials for several other tumor types |
|
Abemaciclib (LY2835219) | D1-CDK4, 0.6 to 2 nM; D3-CDK6, 8 nM |
Cyclin T1–CDK9, PIM1, HIPK2, CDKL5, CAMK2A, CAMK2D, CAMK2G, GSK3α/β, and (at higher doses) cyclin E/A–CDK2 and cyclin B–CDK1 |
FDA-approved for early (adjuvant) and advanced HR+/HER2– breast cancer in combination with endocrine therapy; FDA-approved as monotherapy in advanced HR+/HER2– breast cancer; phase 2/3 trials for several other tumor types |
Trilaciclib (G1T28) | D1-CDK4, 1 nM; D3-CDK6, 4 nM |
FDA-approved for small-cell lung cancer to reduce chemotherapy-induced bone marrow suppression; phase 2/3 trials for other solid tumors |
|
Lerociclib (G1T38) | D1-CDK4, 1 nM; D3-CDK6, 2 nM |
Phase 1/2 trials for HR+/HER2– advanced breast cancer and EGFR-mutant non–small-cell lung cancer |
|
SHR6390 | CDK4, 12 nM; CDK6, 10 nM |
Phase 1/2/3 trials for HR+/HER2– advanced breast cancer and other solid tumors |
|
PF-06873600 | CDK4, 0.13 nM (Ki), CDK6, 0.16 nM (Ki) |
CDK2, 0.09 nM (Ki) | Phase 2 trials for HR+/HER2– advanced breast cancer and other solid tumors |
FCN-437 | D1-CDK4, 3.3 nM; D3-CDK6, 13.7 nM |
Phase 1/2 trials for HR+/HER2– advanced breast cancer and other solid tumors |
|
Birociclib (XZP-3287) | Not reported | Phase 1/2 trials for HR+/HER2– advanced breast cancer and other solid tumors |
|
HS-10342 | Not reported | Phase 1/2 trials for HR+/HER2– advanced breast cancer and other solid tumors |
|
CS3002 | Not reported | Phase 1 trial for solid tumors |
Validation of CDK4/6 inhibitors as anticancer agents
Tumor cell senescence upon CDK4/6 inhibition
Markers predicting response to CDK4/6 inhibition
Synergy of CDK4/6 inhibitors with other compounds
CDK4/6 inhibitor | Synergistic target | Inhibitor | Disease |
---|---|---|---|
Palbociclib | PI3K | Taselisib, pictilisib | PIK3CA mutant TNBC |
AR | Enzalutamide | Androgen receptor–positive TNBC | |
EGFR | Erlotinib | TNBC, esophageal squamous cell carcinoma | |
RAF | PLX4720 | BRAF-V600E mutant melanoma | |
MEK | Trametinib | KRAS mutant colorectal cancer | |
MEK | PD0325901 (mirdametinib) | KRAS or BRAFV600E mutant colorectal cancer | |
MEK | MEK162 (binimetinib) | KRAS mutant colorectal cancer | |
MEK | AZD6244 (selumetinib) | Pancreatic ductal adenocarcinoma | |
PI3K/mTOR | BEZ235 (dactolisib), AZD0855, GDC0980 (apitolisib) | Pancreatic ductal adenocarcinoma | |
IGF1R/InsR | BMS-754807 | Pancreatic ductal adenocarcinoma | |
mTOR | Temsirolimus | Pancreatic ductal adenocarcinoma | |
mTOR | AZD2014 (vistusertib) | ER+ breast cancer | |
mTOR | MLN0128 (sapanisertib) | Intrahepatic cholangiocarcinoma | |
mTOR | Everolimus | Melanoma, glioblastoma | |
Ribociclib | PI3K | GDC-0941 (pictilisib), BYL719 (alpelisib) | PIK3CA mutant breast cancer |
PDK1 | GSK2334470 | ER+ breast cancer | |
EGFR | Nazartinib | EGFR-mutant lung cancer | |
RAF | Encorafenib | BRAF-V600E mutant melanoma | |
mTOR | Everolimus | T-ALL | |
Inflammation | Glucocorticoid dexamethasone | T-ALL | |
γ-Secretase | Compound E | T-ALL | |
Abemaciclib | HER2 | Trastuzumab | HER2+ breast cancer |
EGFR and HER2 | Lapatinib | HER2+ breast cancer | |
RAF | LY3009120, vemurafenib | KRAS mutant lung or colorectal cancer, NRAS or BRAF-V600E mutant melanoma |
|
Temozolomide (alkylating agent) | Glioblastoma |
Protection against chemotherapy-induced toxicity
Metabolic function of CDK4/6 in cancer cells

CDK4/6 inhibitors and antitumor immune responses
CDK4/6 inhibitors in clinical trials
CDK4/6 inhibitor |
Trial name | Trial details | Treatment | Patients | Outcome | Ref. | Other outcomes |
---|---|---|---|---|---|---|---|
Palbociclib | PALOMA-1 | Randomized phase 2 |
Aromatase inhibitor letrozole alone (standard of care) versus letrozole plus palbociclib |
Postmenopausal women with advanced ER+/HER2– breast cancer who had not received any systemic treatment for their advanced disease |
Addition of palbociclib markedly increased median PFS from 10.2 months in the letrozole group to 20.2 months in the palbociclib plus letrozole group |
(78) | On the basis of this result, palbociclib received a “Breakthrough Therapy” designation status from FDA and was granted accelerated approval, in combination with letrozole, for the treatment of ER+/HER2– metastatic breast cancer |
Palbociclib | PALOMA-2 | Double-blind phase 3 |
Palbociclib plus letrozole as first- line therapy |
Postmenopausal women with ER+/HER2– breast cancer |
Addition of palbociclib strongly increased median PFS: 14.5 months in the placebo- letrozole group versus 24.8 months in the palbociclib-letrozole group |
(123) | Palbociclib was equally efficacious in patients with luminal A and B breast cancers, and there was no single biomarker associated with the lack of clinical benefit, except for RB1 loss; CDK4 amplification was associated with endocrine resistance, but this was mitigated by addition of palbociclib; tumors with high levels of FGFR2 and ERBB3 mRNA displayed greater PFS gain after addition of palbociclib (79) |
Palbociclib | PALOMA-3 | Randomized phase 3 |
Estrogen receptor antagonist fulvestrant plus placebo versus fulvestrant plus palbociclib |
Women with HR+/HER2– metastatic breast cancer that had progressed on previous endocrine therapy |
The study demonstrated a substantial prolongation of median PFS in the palbociclib- treated group: 4.6 months in the placebo plus fulvestrant group versus 9.5 months in the palbociclib plus fulvestrant group; addition of palbociclib also extended median overall survival from 28.0 months (placebo-fulvestrant) to 34.9 months (palbociclib- fulvestrant); estimated rate of survival at 3 years was 41% versus 50%, respectively |
(124, 125, 135) | |
Palbociclib | NeoPalAna | Palbociclib in an neoadjuvant setting (i.e., prior to surgery) |
Compared the effects of an aromatase inhibitor anastrozole versus palbociclib plus anastrozole on tumor cell proliferation |
Women with newly diagnosed clinical stage II/III ER+/HER2– breast cancer |
Addition of palbociclib enhanced the antiproliferative effect of anastrozole |
(161) | |
Palbociclib | PALLAS | Randomized phase 3 |
Palbociclib plus standard endocrine therapy versus endocrine therapy alone |
Patients with early (stage 2 or 3), HR+/HER2– breast cancer |
Preliminary results indicate that the trial is unlikely to show a statistically significant improvement of invasive disease-free survival |
(138) | |
Palbociclib | PENELOPE-B | Palbociclib in patients with early breast cancer at high risk of recurrence |
Ongoing | ||||
Ribociclib | MONA LEESA-2 |
Randomized phase 3 |
Ribociclib plus letrozole versus placebo plus letrozole |
First-line treatment for postmenopausal women with HR+/HER2– recurrent or metastatic breast cancer who had not received previous systemic therapy for advanced disease |
At 18 months, PFS was 42.2% in the placebo-letrozole group and 63.0% in the ribociclib- letrozole group |
(126) | |
Ribociclib | MONA LEESA-3 |
Phase 3 | Ribociclib plus fulvestrant |
Patients with advanced (metastatic or recurrent) HR+/HER2– breast cancer who have either received no treatment for the advanced disease or previously received a single line of endocrine therapy for the advanced disease |
Addition of ribociclib significantly extended median PFS, from 12.8 months (placebo-fulvestrant) to 20.5 months (ribociclib- fulvestrant); overall survival at 42 months was also extended from 45.9% (placebo-fulvestrant) to 57.8% (ribociclib-fulvestrant) |
(127, 133) | |
Ribociclib | MONA LEESA-7 |
Phase 3 randomized, double-blind |
Ribociclib versus placebo together with an anti- estrogen tamoxifen or an aromatase inhibitor (letrozole or anastrozole) |
Premenopausal and perimenopausal women with HR+/HER2– advanced breast cancer who had not received previous treatment with CDK4/6 inhibitors |
Ribociclib significantly increased median PFS from 13.0 months in the placebo-endocrine therapy group to 23.8 months in the ribociclib-endocrine therapy group; overall survival was also strongly prolonged in the ribociclib group (estimated overall survival at 42 months was 46.0% for the placebo group and 70.2% in the ribociclib group) |
(128, 132) | |
Ribociclib | EarLEE-1 | Phase 3 trial | Ribociclib in the treatment of early- stage, high-risk HR+/HER2– breast cancers |
Ongoing | |||
Abemaciclib | MONARCH 1 | Phase 2 trial | Abemaciclib as a single agent |
Women with HR+/HER2– metastatic breast cancer who had progressed on or after prior endocrine therapy and had 1 or 2 chemotherapy regimens in the metastatic setting |
Abemaciclib exhibited promising activity in these heavily pretreated patients with poor prognosis; median PFS was 6.0 months and overall survival 17.7 months |
(136) | The most common adverse events were diarrhea, fatigue, and nausea (136) |
Abemaciclib | MONARCH 2 | Double-blind phase 3 |
Abemaciclib in combination with fulvestrant |
Women with HR+/HER2– breast cancer who had progressed while receiving endocrine therapy, or while receiving first-line endocrine therapy for metastatic disease |
Addition of abemaciclib significantly increased PFS from 9.3 months in the placebo-fulvestrant to 16.4 in the abemaciclib-fulvestrant group; median overall survival was also extended from 37.3 months to 46.7 months |
(129, 134) | |
Abemaciclib | MONARCH 3 | Randomized phase 3 double-blind |
Abemaciclib plus an aromatase inhibitor (anastrozole or letrozole) |
Postmenopausal women with advanced HR+/HER2– breast cancer who had no prior systemic therapy in the advanced setting |
Addition of abemaciclib prolonged PFS from 14.8 months (in the placebo-aromatase inhibitor group) to 28.2 months (abemaciclib-aromatase inhibitor group) |
(130, 131) | |
Abemaciclib | MonarchE | Phase 3 study | Endocrine with or without abemaciclib |
Patients with HR+/HER2– lymph node–positive, high-risk early breast cancer |
Preliminary analysis indicates that addition of abemaciclib resulted in a significant improvement of invasive disease-free survival and of distant relapse- free survival |
(137) | |
Trilaciclib | Randomized phase 2 study |
Chemotherapy alone (gemcitabine and carboplatin), versus concurrent administration of trilaciclib plus chemotherapy, versus administration of trilaciclib prior to chemotherapy (to mitigate the cytotoxic effect of chemotherapy on bone marrow) |
Patients with recurrent or metastatic triple-negative breast cancer who had no more than two previous lines of chemotherapy |
Addition of trilaciclib did not offer detectable myeloprotection, but resulted in increased overall survival (from 12.8 months in the chemotherapy-only group to 20.1 months in the concurrent trilaciclib and chemotherapy group and 17.8 months in trilaciclib before chemotherapy group) |
(162) | The most common adverse events were neutropenia, thrombocytopenia, and anemia (162) |
Palbociclib
Ribociclib
Abemaciclib
Additional target | Inhibitor | Immune checkpoint inhibitor |
Tumor type |
Trial identifier |
---|---|---|---|---|
Palbociclib | ||||
Aromatase | Letrozole, anastrozole, exemestane |
HR+ breast cancer, HR+ ovarian cancer, metastatic breast cancer, metastatic endometrial cancer |
NCT04130152, NCT03054363, NCT03936270, NCT04047758, NCT02692755, NCT02806050, NCT03870919, NCT02040857, NCT04176354, NCT02028507, NCT03220178, NCT02592083, NCT02603679, NCT04256941, NCT03425838, NCT02894398, NCT02297438, NCT02730429, NCT02142868, NCT02942355 |
|
LHRH | LHRH agonists: goserelin, leuprolide |
HR+ breast cancer | NCT03969121, NCT03423199, NCT01723774, NCT02917005, NCT02592746, NCT03628066 |
|
ER | ER antagonists: fulvestrant, tamoxifen |
HR+ breast cancer, metastatic breast cancer |
NCT02668666, NCT02738866, NCT03184090, NCT04526028, NCT02513394, NCT03560856, NCT02760030, NCT03079011, NCT03227328, NCT03809988, NCT02764541, NCT03007979, NCT03633331 |
|
ER | Selective estrogen receptor degraders (SERDs): G1T48, ZN-c5, SAR439859, AZD9833, GDC-9545 |
HR+ breast cancer | NCT03455270, NCT04546009, NCT04436744, NCT04478266, NCT03560531, NCT03616587, NCT03284957, NCT03332797 |
|
ER | Selective estrogen receptor modulator (SERM): bazedoxifene |
HR+ breast cancer | NCT03820830, NCT02448771 |
|
Aromatase + PD-1 | Letrozole, anastrozole | Pembrolizumab, nivolumab |
Stage IV ER+ breast cancer |
NCT02778685, NCT04075604 |
PD-1 | Nivolumab, pembrolizumab, MGA012 |
Liposarcoma | NCT04438824 | |
PD-L1 | Avelumab | AR+ breast cancer, TNBC, ER+/HER2– metastatic breast cancer |
NCT04360941, NCT03147287 |
|
EGFR + PD-L1 | Cetuximab | Avelumab | Squamous cell carcinoma of the head and neck |
NCT03498378 |
HER2 | Tucatinib, trastuzumab, pertuzumab, T-DM1, ZW25 |
HER2+ breast cancer | NCT03530696, NCT03054363, NCT02448420, NCT03709082, NCT03304080, NCT02947685 |
|
EGFR/HER2 | Neratinib | Advanced solid tumors with EGFR mutation/amplification, HER2 mutation/amplification, HER3/4 mutation, or KRAS mutation |
NCT03065387 | |
EGFR | Cetuximab | Metastatic colorectal cancer, squamous cell carcinoma of the head and neck |
NCT03446157, NCT02499120 |
|
FGFR | Erdafitinib | ER+/HER2–/FGFR-amplified metastatic breast cancer |
NCT03238196 | |
FGFR1-3 | Rogaratinib | FGFR1-3+/HR+ breast cancer | NCT04483505 | |
IGF-1R | Ganitumab | Ewing sarcoma | NCT04129151 | |
VEGF1-3 receptors + PD-L1 |
Axitinib | Avelumab | NSCLC | NCT03386929 |
RAF | Sorafenib | Leukemia | NCT03132454 | |
MEK | PD-0325901, binimetinib |
KRAS mutant NSCLC, TNBC, KRAS and NRAS mutant metastatic or unresectable colorectal cancer |
NCT02022982, NCT03170206, NCT04494958, NCT03981614 |
|
ERK | Ulixertinib | Advanced pancreatic cancer and other solid tumors |
NCT03454035 | |
PI3K | Copanlisib | HR+ breast cancer | NCT03128619 | |
PI3K | Taselisib, pictilisib, GDC-0077 |
PIK3CA mutant advanced solid tumors, PIK3CA mutant and HR+ breast cancer |
NCT02389842, NCT04191499, NCT03006172 |
|
PI3K/mTOR | Gedatolisib | Metastatic breast cancer, advanced squamous cell lung, pancreatic, head and neck cancer and other solid tumors |
NCT02684032, NCT03065062, NCT02626507 |
|
mTOR | Everolimus, vistusertib | HR+ breast cancer | NCT02871791 | |
AKT | Ipatasertib | HR+ breast cancer, metastatic breast cancer, metastatic gastrointestinal tumors, NSCLC |
NCT03959891, NCT04060862, NCT04591431 |
|
BTK | Ibrutinib | Mantle cell lymphoma | NCT03478514 | |
BCL-2 | Venetoclax | ER+/BCL-2+ advanced or metastatic breast cancer |
NCT03900884 | |
AR | AR antagonists: bicalutamide | AR+ metastatic breast cancer | NCT02605486 | |
Lysosome + aromatase |
Hydroxychloroquine + letrozole | ER+ breast cancer | NCT03774472 | |
Proliferating cells | Standard chemotherapy | Stage IV ER+ breast cancer | NCT03355157 | |
Proliferating cells | Radiation | Stage IV ER+ breast cancer | NCT03870919, NCT03691493, NCT04605562 |
|
BCR-ABL | Bosutinib | HR+ breast cancer | NCT03854903 | |
Ribociclib | ||||
Aromatase | Letrozole, anastrozole, exemestane |
HR+ breast cancer, metastatic breast cancer, ovarian cancer |
NCT04256941, NCT03425838, NCT03822468, NCT02712723, NCT03673124, NCT02941926, NCT03248427, NCT03671330, NCT02333370, NCT01958021, NCT03425838 |
|
LHRH | LHRH agonists: goserelin, leuprolide |
HR+ breast cancer | NCT03944434 | |
ER | ER antagonists: fulvestrant | HR+ breast cancer, advanced breast cancer |
NCT03227328, NCT02632045, NCT02632045, NCT03555877 |
|
PD-1 | Spartalizumab | Breast cancer and ovarian cancer, recurrent and/or metastatic head and neck squamous cell carcinoma, melanoma |
NCT03294694, NCT04213404, NCT03484923 |
|
HER2 | Trastuzumab, pertuzumab, T-DM1 |
HER2+ breast cancer | NCT03913234, NCT02657343 |
|
EGFR | Nazartinib (EGF816) | EGFR mutant NSCLC | NCT03333343 | |
RAF | Encorafenib, LXH254 | NSCLC, BRAF mutant melanoma |
NCT02974725, NCT03333343, NCT04417621, NCT02159066 |
|
MEK | Binimetinib | BRAF V600-dependent advanced solid tumors, melanoma |
NCT01543698, NCT02159066 |
|
PI3K | Alpelisib | Breast cancer with PIK3CA mutation |
NCT03439046 | |
mTOR | Everolimus | Advanced dedifferentiated liposarcoma, leiomyosarcoma, glioma, astrocytoma, glioblastoma, endometrial carcinoma, pancreatic cancer, neuroendocrine tumors |
NCT03114527, NCT03355794, NCT03834740, NCT03008408, NCT02985125, NCT03070301 |
|
mTOR + inflammation | Everolimus + dexamethasone | ALL | NCT03740334 | |
SHP2 | TNO155 | Advanced solid tumors | NCT04000529 | |
AR | AR antagonists: bicalutamide, enzalutamide |
TNBC, metastatic prostate carcinoma |
NCT03090165, NCT02555189 |
|
HDAC | Belinostat | TNBC, ovarian cancer | NCT04315233 | |
proliferating cells | Standard chemotherapy | Ovarian cancer, metastatic solid tumors, soft tissue sarcoma, hepatocellular carcinoma |
NCT03056833, NCT03237390, NCT03009201, NCT02524119 |
|
Abemaciclib | ||||
Aromatase | Letrozole, anastrozole, exemestane |
HR+ breast cancer, metastatic breast cancer, endometrial cancer |
NCT04256941, NCT03425838, NCT04227327, NCT04393285, NCT04305236, NCT03643510, NCT03675893, NCT04352777, NCT04293393, NCT02057133 |
|
ER | ER antagonists: fulvestrant | Advanced breast cancer, low-grade serous ovarian cancer |
NCT03227328, NCT03531645, NCT04158362, NCT01394016 |
|
PD-1 | Nivolumab, pembrolizumab |
Head and neck cancer, g astroesophageal cancer, NSCLC, HR+ breast cancer |
NCT04169074, NCT03655444, NCT03997448, NCT02779751 |
|
ER + PD-L1 | ER antagonists: fulvestrant | Atezolizumab | HR+ breast cancer, metastatic breast cancer |
NCT03280563 |
AKT + ER + PD-L1 | Ipatasertib + ER antagonists: fulvestrant |
Atezolizumab | HR+ breast cancer | NCT03280563 |
PD-L1 | LY3300054 | Advanced solid tumors | NCT02791334 | |
HER2 | Trastuzumab | HER2+ metastatic breast cancer |
NCT04351230 | |
Receptor tyrosine kinases |
Sunitinib | Metastatic renal cell carcinoma |
NCT03905889 | |
IGF-1/IGF-2 | Xentuzumab | HR+ breast cancer | NCT03099174 | |
VEGF-A | Bevacizumab | Glioblastoma | NCT04074785 | |
PI3K | Copanlisib | HR+ breast cancer, metastatic breast cancer |
NCT03939897 | |
PI3K/mTOR | LY3023414 | Metastatic cancer | NCT01655225 | |
ERK1/2 | LY3214996 | tumors with ERK1/2 mutations, glioblastoma, metastatic cancer |
NCT04534283, NCT04391595, NCT02857270 |
|
Trilaciclib | ||||
Proliferating cells | Chemotherapy | SCLC: This trial evaluates the potential clinical benefit of trilaciclib in preventing chemotherapy-induced myelosuppression in patients receiving chemotherapy |
NCT04504513 | |
Proliferating cells + PD-L1 |
Carboplatin + etoposide | Atezolizumab | SCLC: This trial investigates the potential clinical benefit of trilaciclib in preserving the bone marrow and the immune system, and enhancing antitumor efficacy when administered with chemotherapy |
NCT03041311 |
Proliferating cells | Topotecan | SCLC: This trial investigates the potential clinical benefit of trilaciclib in preserving the bone marrow and the immune system, and enhancing the antitumor efficacy of chemotherapy when administered prior to chemotherapy |
NCT02514447 | |
Proliferating cells | Carboplatin + gemcitabine | Metastatic TNBC: This study investigates the potential clinical benefit of trilaciclib in preserving the bone marrow and the immune system, and enhancing the antitumor efficacy of chemotherapy when administered prior to chemotherapy |
NCT02978716 | |
Lerociclib | ||||
ER | ER antagonist: fulvestrant | HR+/HER2– metastatic breast cancer |
NCT02983071 | |
EGFR | Osimertinib | EGFR mutant NSCLC | NCT03455829 | |
SHR6390 | ||||
ER | ER antagonist: fulvestrant | HR+/HER2– recurrent/ metastatic breast cancer |
NCT03481998 | |
Aromatase | Letrozole, anastrozole | HR+/HER2– recurrent/ metastatic breast cancer |
NCT03966898, NCT03772353 |
|
EGFR/HER2 | Pyrotinib | HER2+ gastric cancer, HER2+ metastatic breast cancer |
NCT04095390, NCT03993964 |
|
AR | AR antagonists: SHR3680 | metastatic TNBC | NCT03805399 | |
PF-06873600 | ||||
Endocrine therapy | Single agent and then in combination with endocrine therapy |
HR+/HER2– metastatic breast cancer, ovarian and fallopian tube cancer, TNBC and other tumors |
NCT03519178 | |
FCN-473c | ||||
Aromatase | Letrozole | ER+/HER2– advanced breast cancer |
NCT04488107 |
Resistance to CDK4/6 inhibitors

Outlook
CDK inhibitors and Breast Cancer
The U.S. Food and Drug Administration today granted accelerated approval to Ibrance (palbociclib) to treat advanced (metastatic) breast cancer inr postmenopausal women with estrogen receptor (ER)-positive, human epidermal growth factor receptor 2 (HER2)-negative metastatic breast cancer who have not yet received an endocrine-based therapy. It is to be used in combination with letrozole, another FDA-approved product used to treat certain kinds of breast cancer in postmenopausal women.
See Dr. Melvin Crasto’s blog posts on the announcement of approval of Ibrance (palbociclib) at
and about the structure and mechanism of action of palbociclib
http://newdrugapprovals.org/2014/01/05/palbociclib/
From the CancerNetwork at http://www.cancernetwork.com/aacr-2014/cdk-inhibitors-show-impressive-activity-advanced-breast-cancer
CDK Inhibitors Show Impressive Activity in Advanced Breast Cancer
News | April 08, 2014 | AACR 2014, Breast Cancer
Chemical structure of palbociclib
Palbociclib and LY2835219 are both cyclin-dependent kinase (CDK) 4/6 inhibitors. CDK4 and CDK6 are kinases that, together with cyclin D1, facilitate the transition of dividing cells from the G1 to the S (synthesis) phase of the cell cycle. Preclinical studies have shown that breast cancer cells rely on CDK4 and CDK6 for division and growth, and that selective CDK4/6 inhibitors can arrest the cells at this G1/S phase checkpoint.
The results of the phase II trial of palbociclib and phase I trial of LY2835219 both indicated that hormone receptor (HR)-positive disease appears to be the best marker to predict patient response.
LY2835219 Phase I Trial Demonstrates Early Activity
The CDK4/6 inhibitor LY2835219 has demonstrated early activity in heavily pretreated women with metastatic breast cancer. Nineteen percent of these women (9 out of 47) had a partial response and 51% (24 out of 47) had stable disease following monotherapy with the oral CDK4/6 inhibitor. Patients had received a median of seven prior therapies, and 75% had metastatic disease in the lung, liver, or brain. The median age of patients was 55 years.
All of the partial responses were in patients with HR-positive disease. The overall response rate for this patient subset was 25% (9 of 36 patients). Twenty of the patients with stable disease had HR-positive disease, with 13 patients having stable disease lasting 24 weeks or more.
Despite treatment, disease progression occurred in 23% of the patients.
These results were presented at a press briefing by Amita Patnaik, MD, associate director of clinical research at South Texas Accelerated Research Therapeutics in San Antonio, Texas, at the 2014 American Association for Cancer Research (AACR) Annual Meeting, held April 5–9, in San Diego.
The phase I trial of LY2835219 enrolled 132 patients with five different tumor types, including metastatic breast cancer. Patients received 150-mg to 200-mg doses of the oral drug every 12 hours.
The overall disease control rate was 70% for all patients and 81% among the 36 HR-positive patients.
The median progression-free survival (PFS) was 5.8 months for all patients and 9.1 months for HR-positive patients. Patnaik noted that the median PFS is still a moving target, as 18 patients, all with HR-positive disease, remain on therapy.
“The data are rather encouraging for a very heavily pretreated patient population,” said Patnaik during the press briefing.
Even though the trial was not designed to compare efficacy based on breast cancer subpopulations, the results in HR-positive tumors are particularly encouraging, according to Patnaik.
Common adverse events thought to be treatment-related were diarrhea, nausea, fatigue, vomiting, and neutropenia. These adverse events occurred in 5% or less of patients at grade 3 or 4 toxicity, except neutropenia, which occurred as a grade 3 or 4 toxicity in 11% of patients. Patnaik noted during the press briefing that the neutropenia was uncomplicated and did not result in discontinuation of therapy by any of the patients.
Palbociclib Phase II Data “Impressive”
The addition of the oral CDK4/6 inhibitor palbociclib resulted in an almost doubling of PFS in first-line treatment of postmenopausal metastatic breast cancer patients with HR-positive disease compared with a control population. The patients in this trial were not previously treated for their metastatic breast cancer, unlike the patient population in the phase I LY2835219 trial.
Patients receiving the combination of palbociclib at 125 mg once daily plus letrozole at 2.5 mg once daily had a median PFS of 20.2 months compared with a median of 10.2 months for patients treated with letrozole alone (hazard ratio = 0.488; P = .0004).
Richard S. Finn, MD, assistant professor of medicine at the University of California, Los Angeles, presented the data from the phase II PALOMA-1 trial at a press briefing at the AACR Annual Meeting.
A total of 165 patients were randomized 1:1 to either the experimental arm or control arm.
Forty-three percent of patients in the combination arm had an objective response compared with 33% of patients in the control arm.
Overall survival (OS), a secondary endpoint in this trial, was encouraging but the results are still preliminary, said Finn during the press briefing. The median OS was 37.5 months in the palbociclib arm compared with 33.3 months in the letrozole alone arm (P = .21). Finn noted that long-term follow-up is necessary to establish the median OS. “This first look of the survival data is encouraging. This is a front-line study, and it is encouraging that there is early [separation] of the curves,” he said.
No new toxicities were reported since the interim trial results. Common adverse events included leukopenia, neutropenia, and fatigue. The neutropenia could be quickly resolved and was uncomplicated and not accompanied by fever, said Finn.
Palbociclib is currently being tested in two phase III clinical trials: The PALOMA-3 trial is testing the combination of palbociclib with letrozole and fulvestrant in late-stage metastatic breast cancer patients who have failed endocrine therapy. The PENELOPE-B trial is testing palbociclib in combination with standard endocrine therapy in HR-positive breast cancer patients with residual disease after neoadjuvant chemotherapy and surgery.
References
- Patnaik A, Rosen LS, Tolaney SM, et al. Clinical activity of LY2835219, a novel cell cycle inhibitor selective for CDK4 and CDK6, in patients with metastatic breast cancer. American Association for Cancer Research Annual Meeting 2014; April 5–9, 2014; San Diego. Abstr CT232.
- Finn RS, Crown JP, Lang I, et al. Final results of a randomized phase II study of PD 0332991, a cyclin-dependent kinase (CDK)-4/6 inhibitor, in combination with letrozole vs letrozole alone for first-line treatment of ER+/HER2-advanced breast cancer (PALOMA-1; TRIO-18). American Association for Cancer Research Annual Meeting 2014; April 5–9, 2014; San Diego. Abstr CT101.
– See more at: http://www.cancernetwork.com/aacr-2014/cdk-inhibitors-show-impressive-activity-advanced-breast-cancer#sthash.f29smjxi.dpuf
The Cell Cycle and Anti-Cancer Targets
From Cell Cycle in Cancer: Cyclacel Pharmaceuticals™ (note dotted arrows show inhibition of steps e.g. p21, p53)
For a nice video slideshow explaining a bit more on cyclins and the cell cycle please see video below:
Cell Cycle. 2012 Nov 1; 11(21): 3913.
doi: 10.4161/cc.22390
PMCID: PMC3507481
Cyclin-dependent kinase 4/6 inhibition in cancer therapy
Neil Johnson and Geoffrey I. Shapiro*
See the article “Therapeutic response to CDK4/6 inhibition in breast cancer defined by ex vivo analyses of human tumors” in volume 11 on page 2756.
See the article “CDK4/6 inhibition antagonizes the cytotoxic response to anthracycline therapy” in volume 11 on page 2747.
This article has been cited by other articles in PMC.
Cyclin-dependent kinases (CDKs) drive cell cycle progression and control transcriptional processes. The dysregulation of multiple CDK family members occurs commonly in human cancer; in particular, the cyclin D-CDK4/6-retinoblastoma protein (RB)-INK4 axis is universally disrupted, facilitating cancer cell proliferation and prompting long-standing interest in targeting CDK4/6 as an anticancer strategy. Most agents that have been tested inhibit multiple cell cycle and transcriptional CDKs and have carried toxicity. However, several selective and potent inhibitors of CDK4/6 have recently entered clinical trial. PD0332991, the first to be developed, resulted from the introduction of a 2-aminopyridyl substituent at the C2-position of a pyrido(2,3-d)pyrimidin-7-one backbone, affording exquisite selectivity toward CDK4/6.1 PD0332991 arrests cells in G1 phase by blocking RB phosphorylation at CDK4/6-specfic sites and does not inhibit the growth of RB-deficient cells.2 Phase I studies conducted in patients with advanced RB-expressing cancers demonstrated mild side effects and dose-limiting toxicities of neutropenia and thrombocytopenia, with prolonged stable disease in 25% of patients.3,4 In cyclin D1-translocated mantle cell lymphoma, PD0332991 extinguished CDK4/6 activity in patients’ tumors, resulting in markedly reduced proliferation, and translating to more than 1 year of stability or response in 5 of 17 cases.5
Two recent papers from the Knudsen laboratory make several important observations that will help guide the continued clinical development of CDK4/6 inhibitors. In the study by Dean et al., surgically resected patient breast tumors were grown on a tissue culture matrix in the presence or absence of PD0332991. Crucially, these cultures retained associated stromal components known to play important roles in cancer pathogenesis and therapeutic sensitivities, as well as key histological and molecular features of the primary tumor, including expression of ER, HER2 and Ki-67. Similar to results in breast cancer cell lines,6 the authors demonstrate that only RB-positive tumors have growth inhibition in response to PD0332991, irrespective of ER or HER2 status, while tumors lacking RB were completely resistant. This result underscores RB as the predominant target of CDK4/6 in breast cancer cells and the primary marker of drug response in primary patient-derived tumors. As expected, RB-negative tumors routinely demonstrated robust expression of p16INK4A; however, p16INK4A expression was not always a surrogate marker for RB loss, supporting the importance of direct screening of tumors for RB expression to select patients appropriate for CDK4/6 inhibitor clinical trials.
In the second study, McClendon et al. investigated the efficacy of PD0332991 in combination with doxorubicin in triple-negative breast cancer cell lines. Again, RB functionality was paramount in determining response to either PD0332991 monotherapy or combination treatment. In RB-deficient cancer cells, CDK4/6 inhibition had no effect in either instance. However, in RB-expressing cancer cells, CDK4/6 inhibition and doxorubicin provided a cooperative cytostatic effect, although doxorubicin-induced cytotoxicity was substantially reduced, assessed by markers for mitotic catastrophe and apoptosis. Additionally, despite cytostatic cooperativity, CDK4/6 inhibition maintained the viability of RB-proficient cells in the presence of doxorubicin, which repopulated the culture after removal of drug. These results reflect previous data demonstrating that ectopic expression of p16INK4A can protect cells from the lethal effects of DNA damaging and anti-mitotic chemotherapies.7 Similar results have been reported in MMTV-c-neu mice bearing RB-proficient HER2-driven tumors, where PD0332991 compromised carboplatin-induced regressions,8 suggesting that DNA-damaging treatments should not be combined concomitantly with CDK4/6 inhibition in RB-proficient tumors.
To combine CDK4/6 inhibition with cytotoxics, sequential treatment may be considered, in which CDK4/6 inhibition is followed by DNA damaging chemotherapy; cells relieved of G1 arrest may synchronously enter S phase, where they may be most susceptible to agents disrupting DNA synthesis. Release of myeloma cells from a prolonged PD0332991-mediated G1 block leads to S phase synchronization; interestingly, all scheduled gene expression is not completely restored (including factors critical to myeloma survival such as IRF4), further favoring apoptotic responses to cytotoxic agents.9 Furthermore, in RB-deficient tumors, CDK4/6 inhibitors may be used to maximize the therapeutic window between transformed and non-transformed cells treated with chemotherapy. In contrast to RB-deficient cancer cells, RB-proficient non-transformed cells arrested in G1 in response to PD0332991 are afforded protection from DNA damaging agents, thereby reducing associated toxicities, including bone marrow suppression.8
In summary, the current work provides evidence for RB expression as a determinant of response to CDK4/6 inhibition in primary tumors and highlights the complexity of combining agents targeting the cell cycle machinery with DNA damaging treatments.
Notes
Footnotes
Previously published online: www.landesbioscience.com/journals/cc/article/22390
References
- Toogood PL, et al. J Med Chem. 2005;48:2388–406. doi: 10.1021/jm049354h. [PubMed] [Cross Ref]
- Fry DW, et al. Mol Cancer Ther. 2004;3:1427–38. [PubMed]
- Flaherty KT, et al. Clin Cancer Res. 2012;18:568–76. doi: 10.1158/1078-0432.CCR-11-0509. [PubMed] [Cross Ref]
- Schwartz GK, et al. Br J Cancer. 2011;104:1862–8. doi: 10.1038/bjc.2011.177. [PMC free article] [PubMed] [Cross Ref]
- Leonard JP, et al. Blood. 2012;119:4597–607. doi: 10.1182/blood-2011-10-388298. [PubMed] [Cross Ref]
- Dean JL, et al. Oncogene. 2010;29:4018–32. doi: 10.1038/onc.2010.154. [PubMed] [Cross Ref]
- Stone S, et al. Cancer Res. 1996;56:3199–202. [PubMed]
- Roberts PJ, et al. J Natl Cancer Inst. 2012;104:476–87. doi: 10.1093/jnci/djs002. [PMC free article] [PubMed] [Cross Ref]
- Huang X, et al. Blood. 2012;120:1095–106. doi: 10.1182/blood-2012-03-415984. [PMC free article] [PubMed] [Cross Ref]
Cell Cycle. 2012 Jul 15; 11(14): 2756–2761.
doi: 10.4161/cc.21195
PMCID: PMC3409015
Therapeutic response to CDK4/6 inhibition in breast cancer defined by ex vivo analyses of human tumors
Jeffry L. Dean, 1 , 2 A. Kathleen McClendon, 1 , 2 Theresa E. Hickey, 3 Lisa M. Butler, 3 Wayne D. Tilley, 3 Agnieszka K. Witkiewicz, 4 , 2 ,* and Erik S. Knudsen 1 , 2 ,*
Author information ► Copyright and License information ►
See commentary “Cyclin-dependent kinase 4/6 inhibition in cancer therapy” in volume 11 on page 3913.
This article has been cited by other articles in PMC.
Abstract
To model the heterogeneity of breast cancer as observed in the clinic, we employed an ex vivo model of breast tumor tissue. This methodology maintained the histological integrity of the tumor tissue in unselected breast cancers, and importantly, the explants retained key molecular markers that are currently used to guide breast cancer treatment (e.g., ER and Her2 status). The primary tumors displayed the expected wide range of positivity for the proliferation marker Ki67, and a strong positive correlation between the Ki67 indices of the primary and corresponding explanted tumor tissues was observed. Collectively, these findings indicate that multiple facets of tumor pathophysiology are recapitulated in this ex vivo model. To interrogate the potential of this preclinical model to inform determinants of therapeutic response, we investigated the cytostatic response to the CDK4/6 inhibitor, PD-0332991. This inhibitor was highly effective at suppressing proliferation in approximately 85% of cases, irrespective of ER or HER2 status. However, 15% of cases were completely resistant to PD-0332991. Marker analyses in both the primary tumor tissue and the corresponding explant revealed that cases resistant to CDK4/6 inhibition lacked the RB-tumor suppressor. These studies provide important insights into the spectrum of breast tumors that could be treated with CDK4/6 inhibitors, and defines functional determinants of response analogous to those identified through neoadjuvant studies.
Keywords: ER, PD0332991, breast cancer, cell cycle, ex vivo
Introduction
Breast cancer is a highly heterogeneous disease.1–4 Such heterogeneity is known to influence patient response to both standard of care and experimental therapeutics. In regards to biomarker-driven treatment of breast cancers, it was initially recognized that the presence of the estrogen receptor α (ER) in a fraction of breast cancer cells was associated with the response to tamoxifen and similar anti-estrogenic therapies.5,6 Since this discovery, subsequent marker analyses and gene expression profiling studies have further divided breast cancer into a series of distinct subtypes that harbor differing and often divergent therapeutic sensitivities.1–3 While clearly important in considering the use of several current standard of care therapies, these markers, or molecular sub-types, do not necessarily predict the response to new therapeutic approaches that are currently undergoing clinical development. Thus, there is the continued need for functional analyses of drug response and the definition of new markers that can be used to direct treatment strategies.
Currently, all preclinical cancer models are associated with specific limitations. It is well known that cell culture models lack the tumor microenvironment known to have a significant impact on tumor biology and therapeutic response.7–9 Xenograft models are dependent on the host response for the engraftment of tumor cells in non-native tissues, which do not necessarily recapitulate the nuances of complex tumor milieu.10 In addition, genetically engineered mouse models, while enabling the tumor to develop in the context of the host, can develop tumors that do not mirror aspects of human disease.10 Furthermore, it remains unclear whether any preclinical model truly represents the panoply of breast cancer subtypes that are observed in the clinic. Herein, we utilized a primary human tumor explant culture approach to interrogate drug response, as well as specific determinants of therapeutic response, in an unselected series of breast cancer cases.
Cell Cycle. 2012 Jul 15; 11(14): 2747–2755.
doi: 10.4161/cc.21127
PMCID: PMC3409014
CDK4/6 inhibition antagonizes the cytotoxic response to anthracycline therapy
- Kathleen McClendon, 1 , † Jeffry L. Dean, 1 , † Dayana B. Rivadeneira, 1 Justine E. Yu, 1 Christopher A. Reed, 1 Erhe Gao, 2 John L. Farber, 3 Thomas Force, 2 Walter J. Koch, 2 and Erik S. Knudsen 1 ,*
Author information ► Copyright and License information ►
See commentary “Cyclin-dependent kinase 4/6 inhibition in cancer therapy” in volume 11 on page 3913.
This article has been cited by other articles in PMC.
Abstract
Triple-negative breast cancer (TNBC) is an aggressive disease that lacks established markers to direct therapeutic intervention. Thus, these tumors are routinely treated with cytotoxic chemotherapies (e.g., anthracyclines), which can cause severe side effects that impact quality of life. Recent studies indicate that the retinoblastoma tumor suppressor (RB) pathway is an important determinant in TNBC disease progression and therapeutic outcome. Furthermore, new therapeutic agents have been developed that specifically target the RB pathway, potentially positioning RB as a novel molecular marker for directing treatment. The current study evaluates the efficacy of pharmacological CDK4/6 inhibition in combination with the widely used genotoxic agent doxorubicin in the treatment of TNBC. Results demonstrate that in RB-proficient TNBC models, pharmacological CDK4/6 inhibition yields a cooperative cytostatic effect with doxorubicin but ultimately protects RB-proficient cells from doxorubicin-mediated cytotoxicity. In contrast, CDK4/6 inhibition does not alter the therapeutic response of RB-deficient TNBC cells to doxorubicin-mediated cytotoxicity, indicating that the effects of doxorubicin are indeed dependent on RB-mediated cell cycle control. Finally, the ability of CDK4/6 inhibition to protect TNBC cells from doxorubicin-mediated cytotoxicity resulted in recurrent populations of cells specifically in RB-proficient cell models, indicating that CDK4/6 inhibition can preserve cell viability in the presence of genotoxic agents. Combined, these studies suggest that while targeting the RB pathway represents a novel means of treatment in aggressive diseases such as TNBC, there should be a certain degree of caution when considering combination regimens of CDK4/6 inhibitors with genotoxic compounds that rely heavily on cell proliferation for their cytotoxic effects.
Click on Video Link for Dr. Tolaney slidepresentation of recent data with CDK4/6 inhibitor trial results https://youtu.be/NzJ_fvSxwGk
Audio and slides for this presentation are available on YouTube: http://youtu.be/NzJ_fvSxwGk
Sara Tolaney, MD, MPH, a breast oncologist with the Susan F. Smith Center for Women’s Cancers at Dana-Farber Cancer Institute, gives an overview of phase I clinical trials and some of the new drugs being tested to treat breast cancer. This talk was originally given at the Metastatic Breast Cancer Forum at Dana-Farber on Oct. 5, 2013.
A great article on current clinical trials and explanation of cdk inhibitors by Sneha Phadke, DO; Alexandra Thomas, MD at the site OncoLive
cdk4/6 inhibitor Ibrance Has Favorable Toxicity and Adverse Event Profile
As discussed in earlier posts and the Introduction to this chapter on Cytotoxic Chemotherapeutics, most anti-cancer drugs developed either to target DNA, DNA replication, or the cell cycle usually have similar toxicity profile which can limit their therapeutic use. These toxicities and adverse events usually involve cell types which normally exhibit turnover in the body, such as myeloid and lymphoid and granulocytic series of blood cells, epithelial cells lining the mucosa of the GI tract, as well as follicular cells found at hair follicles. This understandably manifests itself as common toxicities seen with these types of agents such as the various cytopenias in the blood, nausea vomiting diarrhea (although there are effects on the chemoreceptor trigger zone), and alopecia.
It was felt that the cdk4/6 inhibitors would show serious side effects similar to other cytotoxic agents and this definitely may be the case as outlined below:
(Side effects of palbociclib) From navigatingcancer.com
Palbociclib may cause side effects. Tell your doctor if any of these symptoms are severe or do not go away:
- nausea
- diarrhea
- vomiting
- decreased appetite
- tiredness
- numbness or tingling in your arms, hands, legs, and feet
- sore mouth or throat
- unusual hair thinning or hair loss
Some side effects can be serious. If you experience any of these symptoms, call your doctor immediately or get emergency medical treatment:
- fever, chills, or signs of infection
- shortness of breath
- sudden, sharp chest pain that may become worse with deep breathing
- fast, irregular, or pounding heartbeat
- rapid breathing
- weakness
- unusual bleeding or bruising
- nosebleeds
The following is from FDA Drug Trials Snapshot of Ibrance™:
See PDF on original submission and CDER review
original FDA Ibrance submission
original FDA Ibrance submission
4.3 Preclinical Pharmacology/Toxicology
For full details, please see Pharmacology/Toxicology review by Dr. Wei Chen The nonclinical studies adequately support the safety of oral administration of palbociclib for the proposed indication and the recommendation from the team is for approval. Non-clinical studies of palbociclib included safety pharmacology studies, genotoxicity
studies, reproductive toxicity studies, pharmacokinetic studies, toxicokinetic studies and repeat-dose general toxicity studies which were conducted in rats and dogs. The pivotal toxicology studies were conducted in compliance with Good Laboratory Practice regulation.
Pharmacology:
As described above, palbociclib is an inhibitor of CDK4 and CDK6. Palbociclib modulates downstream targets of CDK4 and CDK6 in vitro and induces G1 phase cell cycle arrest and therefore acts to inhibit DNA synthesis and cell proliferation. Combination of palbociclib with anti-estrogen agents demonstrated synergistic inhibition
of cell proliferation in ER+ breast cancer cells. Palbociclib showed anti-tumor efficacy in animal tumor model studies. Safety pharmacology studies with palbociclib demonstrated adverse effects on both the respiratory and cardiovascular function of dogs at a dose of 125mg/day (four times and 50-times the human clinical exposure
respectively) based on mean unbound Cmax.
General toxicology:
Palbociclib was studied in single dose toxicity studies and repeated dose studies in rats and dogs. Adverse effects in the bone marrow, lymphoid tissues, and male reproductive organs were observed at clinically relevant exposures. Partial to complete reversibility of toxicities to the hematolymphopoietic and male reproductive systems was demonstrated following a recovery period (4-12 weeks), with the exception of the male reproductive organ findings in dogs. Gastrointestinal, liver, kidney, endocrine/metabolic (altered glucose metabolism), respiratory, ocular, and adrenal effects were also seen.
Genetic toxicology:
Palbociclib was evaluated for potential genetic toxicity in in vitro and in vivo studies. The Ames bacterial mutagenicity assay in the presence or absence of metabolic activation demonstrated non-mutagenicity. In addition, palbociclib did not induce chromosomal aberrations in cultured human peripheral blood lymphocytes in the presence or absence of metabolic activation. Palbociclib was identified as aneugenic based on kinetochore analysis of micronuclei formation in an In vitro assay in CHO-WBL cells. In addition, palbociclib was shown to induce micronucleus formation in male rats at doses 100
mg/kg/day (10x human exposure at the therapeutic dose) in an in vivo rat micronucleus assay.
Reproductive toxicology: No effects on estrous cycle and no reproductive toxicities were noticed in standard assays.
Pharmacovigilance (note please see PDF for more information)
Deaths Associated With Trials: Although a few deaths occurred during some trials no deaths were attributed to the drug.
Non-Serious Adverse Events:
(note a reviewers comment below concerning incidence of pulmonary embolism is a combination trial with letrazole)
Other article in this Open Access Journal on Cell Cycle and Cancer Include:
Tumor Suppressor Pathway, Hippo pathway, is responsible for Sensing Abnormal Chromosome Numbers in Cells and Triggering Cell Cycle Arrest, thus preventing Progression into Cancer
Nonhematologic Cancer Stem Cells [11.2.3]
New methods for Study of Cellular Replication, Growth, and Regulation
Multiple Lung Cancer Genomic Projects Suggest New Targets, Research Directions for Non-Small Cell Lung Cancer
Proteomics, Metabolomics, Signaling Pathways, and Cell Regulation: a Compilation of Articles in the Journal http://pharmaceuticalintelligence.com
In Focus: Targeting of Cancer Stem Cells
Leave a Reply