1
|
Wilson TK, Zishiri OT. Prostate Cancer: A Review of Genetics, Current Biomarkers and Personalised Treatments. Cancer Rep (Hoboken) 2024; 7:e70016. [PMID: 39410867 PMCID: PMC11480670 DOI: 10.1002/cnr2.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/22/2024] [Accepted: 09/09/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Prostate cancer is the second leading cause of cancer deaths in men, second only to lung cancer. Despite this, diagnosis and prognosis methods remain limited, with effective treatments being few and far between. Traditionally, prostate cancer is initially tested for through a prostate serum antigen (PSA) test and a digital rectum examination (DRE), followed by confirmation through an invasive prostate biopsy. The DRE and biopsy are uncomfortable for the patient, so less invasive, accurate diagnostic tools are needed. Current diagnostic tools, along with genes that hold possible biomarker uses in diagnosis, prognosis and indications for personalised treatment plans, were reviewed in this article. RECENT FINDINGS Several genes from multiple families have been identified as possible biomarkers for disease, including those from the MYC and ETS families, as well as several tumour suppressor genes, Androgen Receptor signalling genes and DNA repair genes. There have also been advances in diagnostic tools, including MRI-targeted and liquid biopsies. Several personalised treatments have been developed over the years, including those that target metabolism-driven prostate cancer or those that target inflammation-driven cancer. CONCLUSION Several advances have been made in prostate cancer diagnosis and treatment, but the disease still grows year by year, leading to more and more deaths annually. This calls for even more research into this disease, allowing for better diagnosis and treatment methods and a better chance of patient survival.
Collapse
Affiliation(s)
- Trevor K. Wilson
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering, and ScienceUniversity of KwaZulu‐NatalDurbanSouth Africa
| | - Oliver T. Zishiri
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering, and ScienceUniversity of KwaZulu‐NatalDurbanSouth Africa
| |
Collapse
|
2
|
Rani A, Pulukuri AJ, Wei J, Dhull A, Dar AI, Sharma R, Mesbahi N, Savoy EA, Yoon H, Wu BJ, Berkman CE, Sharma A. PSMA-Targeted 2-Deoxyglucose-Based Dendrimer Nanomedicine for the Treatment of Prostate Cancer. Biomacromolecules 2024; 25:6164-6180. [PMID: 39164913 DOI: 10.1021/acs.biomac.4c00878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Prostate cancer (PC) is the fifth leading cause of cancer-related deaths among men worldwide. Prostate-specific membrane antigen (PSMA), a molecular target of PC, is clinically used for the treatment and diagnosis of PC using radioligand approaches. However, no PSMA-based chemotherapies have yet been approved by the FDA. Here, we present a novel therapeutic approach using PSMA-targeted 2-deoxyglucose-dendrimer (PSMA-2DG-D) for targeted delivery of a potent tyrosine kinase inhibitor, cabozantinib (Cabo), selectively to PC cells. PSMA-2DG-D demonstrates intracellular localization in PSMA (+) PC cells through PSMA-mediated internalization. This PSMA-specific targeting translates to enhanced efficacy of Cabo compared to the free drug when conjugated to PSMA-2DG-D. Furthermore, systemically administered fluorescently labeled PSMA-2DG-D-Cy5 specifically targets PSMA (+) tumors with minimal off-target accumulation in the PC3-PIP tumor xenograft mouse model. This demonstrates that the PSMA-2DG-D platform is a promising new delivery system for potent chemotherapeutics, where systemic side effects are a significant concern.
Collapse
Affiliation(s)
- Anu Rani
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, Washington 99164, United States
| | - Anunay James Pulukuri
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, Washington 99164, United States
| | - Jing Wei
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| | - Anubhav Dhull
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, Washington 99164, United States
| | - Aqib Iqbal Dar
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, Washington 99164, United States
| | - Rishi Sharma
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, Washington 99164, United States
| | - Nooshin Mesbahi
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, Washington 99164, United States
| | - Emily A Savoy
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, Washington 99164, United States
| | - Hosog Yoon
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, Washington 99164, United States
| | - Boyang Jason Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| | - Clifford E Berkman
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, Washington 99164, United States
| | - Anjali Sharma
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, Washington 99164, United States
| |
Collapse
|
3
|
Brown J, Santini D, Charnley N, Ogareva A, Chisholm A, Jones R. Implications of bone metastasis on response to systemic therapy in patients with advanced renal cell carcinoma: A systematic literature review. Cancer Treat Rev 2024; 129:102792. [PMID: 38972135 DOI: 10.1016/j.ctrv.2024.102792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/07/2024] [Accepted: 06/23/2024] [Indexed: 07/09/2024]
Abstract
INTRODUCTION Bone metastases negatively affect prognosis in patients with advanced renal cell carcinoma (aRCC). We conducted a systematic literature review to identify clinical trial publications including patients with aRCC with and without bone metastases. METHODS The review was conducted according to Preferred Reporting Items for Systematic Reviews and Meta‑Analyses (PRISMA) guidelines and registered with PROSPERO (CRD42022355436). MEDLINE and Embase databases were searched (September 2, 2022) to identify publications reporting efficacy and safety outcomes for patients with/without bone metastasis from clinical trials of systemic RCC therapies. Risk of bias was assessed using Grading of Recommendations Assessment, Development and Evaluation (GRADE). RESULTS Of 526 publications screened, 19 were eligible: seven (from five studies) reported phase 3 trials, six reported phase 2 trials, one reported phase 1b/2 trials, and five were pooled analyses. Five publications reported moderate-quality evidence, while 14 were graded as low- or very low-quality evidence, suggesting a high potential for uncertainty. Five studies reported benefits of investigational therapies versus comparators in patients with and without bone metastases; these studies included cabozantinib, nivolumab, cabozantinib plus nivolumab, and lenvatinib plus pembrolizumab treatment arms. Data were also available for nivolumab plus ipilimumab. Bone metastases were consistently associated with poor prognosis in patients with aRCC. Preliminary data support the hypothesis that therapies targeting pathways implicated in the development of bone metastases may be beneficial, and warrant further investigation. However, data to support treatment decision-making are lacking. CONCLUSION Our findings highlight the need for clinical data to assist in defining the optimal treatment for patients with aRCC and bone metastasis.
Collapse
Affiliation(s)
- Janet Brown
- Division of Clinical Medicine, University of Sheffield, Sheffield, UK.
| | - Daniele Santini
- Medical Oncology A, Policlinico Umberto 1, La Sapienza University of Rome, Italy
| | | | | | | | - Robert Jones
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
4
|
Dhull A, Wei J, Pulukuri AJ, Rani A, Sharma R, Mesbahi N, Yoon H, Savoy EA, Xaivong Vi S, Goody KJ, Berkman CE, Wu BJ, Sharma A. PSMA-targeted dendrimer as an efficient anticancer drug delivery vehicle for prostate cancer. NANOSCALE 2024; 16:5634-5652. [PMID: 38440933 DOI: 10.1039/d3nr06520k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Prostate cancer (PCa) is the second leading cause of cancer-related deaths among men in the United States. Although early-stage treatments exhibit promising 5-year survival rates, the treatment options for advanced stage disease are constrained, with short survival benefits due to the challenges associated with effective and selective drug delivery to PCa cells. Even though targeting Prostate Specific Membrane Antigen (PSMA) has been extensively explored and is clinically employed for imaging and radio-ligand therapy, the clinical success of PSMA-based approaches for targeted delivery of chemotherapies remains elusive. In this study, we combine a generation 4 hydroxy polyamidoamine dendrimer (PD) with irreversible PSMA ligand (CTT1298) to develop a PSMA-targeted nanoplatform (PD-CTT1298) for selective intracellular delivery of potent chemotherapeutics to PCa. PD-CTT1298-Cy5 exhibits a PSMA IC50 in the nanomolar range and demonstrates selective uptake in PSMA (+) PCa cells via PSMA mediated internalization. When systemically administered in a prostate tumor xenograft mouse model, PD-CTT1298-Cy5 selectively targets PSMA (+) tumors with significantly less accumulation in PSMA (-) tumors or upon blocking of the PSMA receptors. Moreover, the dendrimer clears rapidly from the off-target organs limiting systemic side-effects. Further, the conjugation of an anti-cancer agent, cabozantinib to the PSMA-targeted dendrimer translates to a significantly enhanced anti-proliferative activity in vitro compared to the free drug. These findings highlight the potential of PD-CTT1298 nanoplatform as a versatile approach for selective delivery of high payloads of potent chemotherapeutics to PCa, where dose related systemic side-effects are a major concern.
Collapse
Affiliation(s)
- Anubhav Dhull
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Jing Wei
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, USA
| | - Anunay James Pulukuri
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Anu Rani
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Rishi Sharma
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Nooshin Mesbahi
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Hosog Yoon
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Emily A Savoy
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Sylvia Xaivong Vi
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Kenneth John Goody
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Clifford E Berkman
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Boyang Jason Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, USA
| | - Anjali Sharma
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| |
Collapse
|
5
|
Alhazzani K, Alsahli M, Alanazi AZ, Algahtani M, Alenezi AA, Alhoshani A, Alqinyah M, Alhamed AS, Alhosaini K. Augmented antitumor effects of erlotinib and cabozantinib on A549 non-small cell lung cancer: In vitro and in vivo studies. Saudi Pharm J 2023; 31:101756. [PMID: 37705877 PMCID: PMC10495648 DOI: 10.1016/j.jsps.2023.101756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/19/2023] [Indexed: 09/15/2023] Open
Abstract
Non-small cell lung carcinoma is a challenging disease worldwide. This study aims to determine whether combining erlotinib, an epidermal growth factor receptor (EGFR) inhibitor, with cabozantinib, a mesenchymal-epithelial transition factor (c-Met) inhibitor, would have an augmented therapeutic benefit on A549 cells. The combination of erlotinib and cabozantinib (5 µM) inhibited A549 cell viability compared to each monotherapy at ≥ 10 µM as confirmed by the MTT assay. Combination therapy also has a more potent inhibition of cellular migration than monotherapy using the wound-healing assay. Furthermore, mRNA expression analyses for assessing apoptosis, metastasis, and cell cycle-related genes, the results showed that combination therapy significantly inhibits levels of BCL-2, MMP-9, VEGF, and TGF-β while inducing p53, p21, and BAX expression. In terms of oncogenic markers, western blotting analysis showed a significant reduction of BCl-2 expression and elevation in caspase3, p53, and p21 proteins as indicators of cell death via apoptosis. The antitumor in vivo effect of the combination therapy showed significant tumor inhibition compared to monotherapy. In conclusion, combination therapy could be a potential promising strategy to treat non-small cell lung carcinoma.
Collapse
Affiliation(s)
- Khalid Alhazzani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Meshal Alsahli
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed Z Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad Algahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ahmad A Alenezi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ali Alhoshani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed Alqinyah
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah S. Alhamed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Khaled Alhosaini
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
6
|
Solimando AG, Kalogirou C, Krebs M. Angiogenesis as Therapeutic Target in Metastatic Prostate Cancer - Narrowing the Gap Between Bench and Bedside. Front Immunol 2022; 13:842038. [PMID: 35222436 PMCID: PMC8866833 DOI: 10.3389/fimmu.2022.842038] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/21/2022] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis in metastatic castration-resistant prostate cancer (mCRPC) has been extensively investigated as a promising druggable biological process. Nonetheless, targeting angiogenesis has failed to impact overall survival (OS) in patients with mCRPC despite promising preclinical and early clinical data. This discrepancy prompted a literature review highlighting the tumor heterogeneity and biological context of Prostate Cancer (PCa). Narrowing the gap between the bench and bedside appears critical for developing novel therapeutic strategies. Searching clinicaltrials.gov for studies examining angiogenesis inhibition in patients with PCa resulted in n=20 trials with specific angiogenesis inhibitors currently recruiting (as of September 2021). Moreover, several other compounds with known anti-angiogenic properties - such as Metformin or Curcumin - are currently investigated. In general, angiogenesis-targeting strategies in PCa include biomarker-guided treatment stratification - as well as combinatorial approaches. Beyond established angiogenesis inhibitors, PCa therapies aiming at PSMA (Prostate Specific Membrane Antigen) hold the promise to have a substantial anti-angiogenic effect - due to PSMA´s abundant expression in tumor vasculature.
Collapse
Affiliation(s)
- Antonio Giovanni Solimando
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine "G. Baccelli", University of Bari Medical School, Bari, Italy.,Medical Oncology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Charis Kalogirou
- Department of Urology and Pediatric Urology, University Hospital Würzburg, Würzburg, Germany
| | - Markus Krebs
- Department of Urology and Pediatric Urology, University Hospital Würzburg, Würzburg, Germany.,Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
7
|
Choy E, Cote GM, Michaelson MD, Wirth L, Gainor JF, Muzikansky A, Sequist LV, Sullivan RJ, Fidias PM, Shaw A, Heist RS. OUP accepted manuscript. Oncologist 2022; 27:600-606. [PMID: 35524758 PMCID: PMC9256024 DOI: 10.1093/oncolo/oyac083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 03/14/2022] [Indexed: 11/23/2022] Open
Abstract
Bone metastases are often difficult to manage as they can be symptomatic and skeletal-related events (SREs) can contribute to significant morbidity and declines in performance status. We sought to identify a novel medical treatment for bone metastasis by testing the safety and efficacy of cabozantinib in patients with bone metastasis arising from non-breast, non-prostate, malignant solid tumors. Patients were administered cabozantinib as an oral drug starting at 60 mg per day and radiologic measurements were performed at baseline and every 8 weeks. Thirty-seven patients were enrolled. No SREs were observed throughout the study. Twenty patients had disease measurable by Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. Four of 20 had a partial response by RECIST. An additional 12 patients had some decrease in tumor burden with nine of these having a decrease in tumor burden of at least 10% by RECIST. Six of the patients with at least a minor response had sarcoma. Sixteen patients had biomarkers of bone turnover measured before and after treatment. Most of these patients demonstrated decrease in urine and serum N-telopeptide and serum C-telopeptide. However, these changes in biomarkers of bone turnover did not correlate with radiographic changes measured by RECIST. This study demonstrates clinical activity and safety for cabozantinib in heavily pretreated patients with bone metastasis and shows activity for cabozantinib in patients with metastatic sarcoma.
Collapse
Affiliation(s)
- Edwin Choy
- Corresponding author: Edwin Choy, MD, Division of Hematology Oncology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA.
| | - Gregory M Cote
- Division of Hematology Oncology, Massachusetts General Hospital, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - M Dror Michaelson
- Division of Hematology Oncology, Massachusetts General Hospital, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Lori Wirth
- Division of Hematology Oncology, Massachusetts General Hospital, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Justin F Gainor
- Division of Hematology Oncology, Massachusetts General Hospital, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Alona Muzikansky
- Massachusetts General Hospital Biostatistics Center, Boston, MA, USA
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Lecia V Sequist
- Division of Hematology Oncology, Massachusetts General Hospital, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Ryan J Sullivan
- Division of Hematology Oncology, Massachusetts General Hospital, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Panagiotis M Fidias
- Division of Hematology Oncology, Massachusetts General Hospital, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Medical Oncology, Center for Cancer Care, Exeter Hospital, Exeter, NH, USA
| | - Alice Shaw
- Division of Hematology Oncology, Massachusetts General Hospital, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Rebecca S Heist
- Division of Hematology Oncology, Massachusetts General Hospital, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| |
Collapse
|
8
|
Shang R, Song X, Wang P, Zhou Y, Lu X, Wang J, Xu M, Chen X, Utpatel K, Che L, Liang B, Cigliano A, Evert M, Calvisi DF, Chen X. Cabozantinib-based combination therapy for the treatment of hepatocellular carcinoma. Gut 2021; 70:1746-1757. [PMID: 33144318 PMCID: PMC8089119 DOI: 10.1136/gutjnl-2020-320716] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 09/25/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer with limited treatment options. Cabozantinib, an orally bioavailable multikinase inhibitor is now approved by Food and Drug Administration (FDA) for HCC patients. We evaluated the therapeutic efficacy of cabozantinib, either alone or in combination, in vitro and in vivo. DESIGN Human HCC cell lines and HCC mouse models were used to assess the therapeutic efficacy and targeted molecular pathways of cabozantinib, either alone or in combination with the pan-mTOR inhibitor MLN0128 or the checkpoint inhibitor anti-PD-L1 antibody. RESULTS Cabozantinib treatment led to stable disease in c-Met/β-catenin and Akt/c-Met mouse HCC while possessing limited efficacy on Akt/Ras and c-Myc liver tumours. Importantly, cabozantinib effectively inhibited c-MET and ERK activity, leading to decreased PKM2 and increased p21 expression in HCC cells and in c-Met/β-catenin and Akt/c-Met HCC. However, cabozantinib was ineffective in inhibiting the Akt/mTOR cascade. Intriguingly, a strong inhibition of angiogenesis by cabozantinib occurred regardless of the oncogenic drivers. However, cabozantinib had limited impact on other tumour microenvironment parameters, including tumour infiltrating T cells, and did not induce programmed death-ligand 1 (PD-L1) expression. Combining cabozantinib with MLN0128 led to tumour regression in c-Met/β-catenin mice. In contrast, combined treatment with cabozantinib and the checkpoint inhibitor anti-PD-L1 antibody did not provide any additional therapeutic benefit in the four mouse HCC models tested. CONCLUSION c-MET/ERK/p21/PKM2 cascade and VEGFR2-induced angiogenesis are the primary targets of cabozantinib in HCC treatment. Combination therapies with cabozantinib and mTOR inhibitors may be effective against human HCC.
Collapse
Affiliation(s)
- Runze Shang
- Department of Hepatobiliary Surgery, Xijing Hospital, Xian, Shaanxi, China,Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA,Department of General Surgery, The 910 Hospital, Quanzhou, Fujian, China
| | - Xinhua Song
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Pan Wang
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA,Collaborative Innovation Center for Agricultural Product Processing and Nutrition & Health, Beijing Vegetable Research Center, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Yi Zhou
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA,Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xinjun Lu
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA,Department of Hepatic Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Jingxiao Wang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, Beijing, China
| | - Meng Xu
- Department of General Surgery, The Second Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinyan Chen
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Kirsten Utpatel
- Institute of Pathology, University of Regensburg, Regensburg, Bayern, Germany
| | - Li Che
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA,R&D Center, Legend Biotech USA Inc, Piscataway, New Jersey, USA
| | - Binyong Liang
- Hepatic Surgery Center, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Antonio Cigliano
- Institute of Pathology, University of Regensburg, Regensburg, Bayern, Germany
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, Regensburg, Bayern, Germany
| | - Diego F Calvisi
- Institute of Pathology, University of Regensburg, Regensburg, Bayern, Germany
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
9
|
Santoni M, Iacovelli R, Colonna V, Klinz S, Mauri G, Nuti M. Antitumor effects of the multi-target tyrosine kinase inhibitor cabozantinib: a comprehensive review of the preclinical evidence. Expert Rev Anticancer Ther 2021; 21:1029-1054. [PMID: 34445927 DOI: 10.1080/14737140.2021.1919090] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Altered receptor tyrosine kinase (RTK) signaling contributes to tumorigenesis and suppression of immune-mediated destruction of cancer cells. Cabozantinib is an oral tyrosine kinase inhibitor that inhibits several RTKs involved in tumorigenesis, and is approved for the treatment of patients with progressive metastatic medullary thyroid cancer, advanced renal cell carcinoma, and hepatocellular carcinoma that has been previously treated with sorafenib. AREAS COVERED We present an up-to-date evaluation of preclinical evidence for RTK inhibition with cabozantinib, specifically VEGFR, MET, KIT, RET, AXL, FLT3, and associated antitumor effects. Preclinical investigations of cabozantinib in combination with other anticancer drugs are also reviewed. EXPERT OPINION Preclinical evidence shows that cabozantinib has antitumor activity against various cancer cells and exhibits synergy with other anticancer agents, including immune checkpoint inhibitors and hormone receptor or metabolic pathway inhibitors. Further optimization of cabozantinib treatment requires the identification of biomarkers of response and resistance, and exploration of complementary drug targets. Investigation of mechanisms of adaptive resistance, such as epithelial to mesenchymal transition (cancer intrinsic) and immunomodulation by the tumor microenvironment (cancer extrinsic), as well as identification of novel drug targets based on characterization of cancer stem cell metabolomic phenotypes, appear to be promising approaches.
Collapse
Affiliation(s)
- Matteo Santoni
- MD, U.O.C. Medical Oncology, Macerata Hospital, Macerata, Italy
| | - Roberto Iacovelli
- Medical Oncologist, Medical Oncology Unit, Fondazione Policlinico Agostino Gemelli IRCCS, Roma, Italy
| | - Valentina Colonna
- Global Medical Development Director, Global Oncology R&D, Ipsen S.p.A., Milano, Italy
| | - Stephan Klinz
- Senior Director, Translational Medicine & Biomarkers, Ipsen, United States, MA, USA
| | - Giorgio Mauri
- Medical Advisor Oncology, Ipsen S.p.A., Milano, Italy
| | - Marianna Nuti
- Professor, Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
10
|
Iuliani M, Simonetti S, Pantano F, Ribelli G, Di Martino A, Denaro V, Vincenzi B, Russo A, Tonini G, Santini D. Antitumor Effect of Cabozantinib in Bone Metastatic Models of Renal Cell Carcinoma. BIOLOGY 2021; 10:781. [PMID: 34440012 PMCID: PMC8389553 DOI: 10.3390/biology10080781] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND The presence of bone metastases in renal cell carcinoma (RCC) negatively affects patients' survival. Data from clinical trials has highlighted a significant benefit of cabozantinib in bone metastatic RCC patients. Here, we evaluated the antitumor effect of cabozantinib in coculture models of renal cell carcinoma (RCC) and osteoblasts (OBs) to investigate whether and how its antiproliferative activity is influenced by OBs. METHODS Bone/RCC models were generated, coculturing green fluorescent protein (GFP)-tagged Caki-1 and 786-O cells with human primary OBs in a "cell-cell contact" system. RCC proliferation and the OB molecular profile were evaluated after the cabozantinib treatment. RESULTS The Caki-1 cell proliferation increased in the presence of OBs (p < 0.0001), while the 786-O cell growth did not change in the coculture with the OBs. The cabozantinib treatment reduced the proliferation of both the Caki-1 (p < 0.0001) and 786-O (p = 0.03) cells cocultured with OBs. Intriguingly, the inhibitory potency of cabozantinib was higher when Caki-1 cells grew in presence of OBs compared to a monoculture (p < 0.001), and this was similar in 786-O cells alone or cocultured with OBs. Moreover, the OB pretreatment with cabozantinib "indirectly" inhibited Caki-1 cell proliferation (p = 0.040) without affecting 786-O cell growth. Finally, we found that cabozantinib was able to modulate the OB gene and molecular profile inhibiting specific proliferative signals that, in turn, could affect RCC cell growth. CONCLUSIONS Overall, the "direct" effect of cabozantinib on OBs "indirectly" increased its antitumor activity in metastatic RCC Caki-1 cells but not in the primary 786-O model.
Collapse
Affiliation(s)
- Michele Iuliani
- Department of Medical Oncology, Campus Bio-Medico University of Rome, 00128 Rome, Italy; (M.I.); (S.S.); (G.R.); (B.V.); (G.T.); (D.S.)
| | - Sonia Simonetti
- Department of Medical Oncology, Campus Bio-Medico University of Rome, 00128 Rome, Italy; (M.I.); (S.S.); (G.R.); (B.V.); (G.T.); (D.S.)
| | - Francesco Pantano
- Department of Medical Oncology, Campus Bio-Medico University of Rome, 00128 Rome, Italy; (M.I.); (S.S.); (G.R.); (B.V.); (G.T.); (D.S.)
| | - Giulia Ribelli
- Department of Medical Oncology, Campus Bio-Medico University of Rome, 00128 Rome, Italy; (M.I.); (S.S.); (G.R.); (B.V.); (G.T.); (D.S.)
| | - Alberto Di Martino
- Department of Biomedical and Neurimotor Sciences (DIBINEM), 1st Orthopaedic Clinic, IRCCS Istituto Ortopedico Rizzoli, 40126 Bologna, Italy;
| | - Vincenzo Denaro
- Department of Orthopaedic and Trauma Surgery, Campus Bio-Medico University of Rome, 00128 Rome, Italy;
| | - Bruno Vincenzi
- Department of Medical Oncology, Campus Bio-Medico University of Rome, 00128 Rome, Italy; (M.I.); (S.S.); (G.R.); (B.V.); (G.T.); (D.S.)
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90133 Palermo, Italy;
| | - Giuseppe Tonini
- Department of Medical Oncology, Campus Bio-Medico University of Rome, 00128 Rome, Italy; (M.I.); (S.S.); (G.R.); (B.V.); (G.T.); (D.S.)
| | - Daniele Santini
- Department of Medical Oncology, Campus Bio-Medico University of Rome, 00128 Rome, Italy; (M.I.); (S.S.); (G.R.); (B.V.); (G.T.); (D.S.)
| |
Collapse
|
11
|
Baca SC, Takeda DY, Seo JH, Hwang J, Ku SY, Arafeh R, Arnoff T, Agarwal S, Bell C, O'Connor E, Qiu X, Alaiwi SA, Corona RI, Fonseca MAS, Giambartolomei C, Cejas P, Lim K, He M, Sheahan A, Nassar A, Berchuck JE, Brown L, Nguyen HM, Coleman IM, Kaipainen A, De Sarkar N, Nelson PS, Morrissey C, Korthauer K, Pomerantz MM, Ellis L, Pasaniuc B, Lawrenson K, Kelly K, Zoubeidi A, Hahn WC, Beltran H, Long HW, Brown M, Corey E, Freedman ML. Reprogramming of the FOXA1 cistrome in treatment-emergent neuroendocrine prostate cancer. Nat Commun 2021; 12:1979. [PMID: 33785741 PMCID: PMC8010057 DOI: 10.1038/s41467-021-22139-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/18/2021] [Indexed: 02/07/2023] Open
Abstract
Lineage plasticity, the ability of a cell to alter its identity, is an increasingly common mechanism of adaptive resistance to targeted therapy in cancer. An archetypal example is the development of neuroendocrine prostate cancer (NEPC) after treatment of prostate adenocarcinoma (PRAD) with inhibitors of androgen signaling. NEPC is an aggressive variant of prostate cancer that aberrantly expresses genes characteristic of neuroendocrine (NE) tissues and no longer depends on androgens. Here, we investigate the epigenomic basis of this resistance mechanism by profiling histone modifications in NEPC and PRAD patient-derived xenografts (PDXs) using chromatin immunoprecipitation and sequencing (ChIP-seq). We identify a vast network of cis-regulatory elements (N~15,000) that are recurrently activated in NEPC. The FOXA1 transcription factor (TF), which pioneers androgen receptor (AR) chromatin binding in the prostate epithelium, is reprogrammed to NE-specific regulatory elements in NEPC. Despite loss of dependence upon AR, NEPC maintains FOXA1 expression and requires FOXA1 for proliferation and expression of NE lineage-defining genes. Ectopic expression of the NE lineage TFs ASCL1 and NKX2-1 in PRAD cells reprograms FOXA1 to bind to NE regulatory elements and induces enhancer activity as evidenced by histone modifications at these sites. Our data establish the importance of FOXA1 in NEPC and provide a principled approach to identifying cancer dependencies through epigenomic profiling.
Collapse
Affiliation(s)
- Sylvan C Baca
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- The Eli and Edythe L. Broad Institute, Cambridge, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David Y Takeda
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Ji-Heui Seo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Justin Hwang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sheng Yu Ku
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rand Arafeh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Taylor Arnoff
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Supreet Agarwal
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Connor Bell
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Edward O'Connor
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Xintao Qiu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sarah Abou Alaiwi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rosario I Corona
- Department of Obstetrics and Gynecology and the Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Marcos A S Fonseca
- Department of Obstetrics and Gynecology and the Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Claudia Giambartolomei
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Istituto Italiano di Tecnologia, Genova, Italy
| | - Paloma Cejas
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Klothilda Lim
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Monica He
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Anjali Sheahan
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Amin Nassar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jacob E Berchuck
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Lisha Brown
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Holly M Nguyen
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Ilsa M Coleman
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Arja Kaipainen
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Navonil De Sarkar
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Peter S Nelson
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Keegan Korthauer
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mark M Pomerantz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Leigh Ellis
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Bogdan Pasaniuc
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Kate Lawrenson
- Department of Obstetrics and Gynecology and the Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kathleen Kelly
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Amina Zoubeidi
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - William C Hahn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- The Eli and Edythe L. Broad Institute, Cambridge, MA, USA
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Henry W Long
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Myles Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Matthew L Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- The Eli and Edythe L. Broad Institute, Cambridge, MA, USA.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
12
|
Labrecque MP, Brown LG, Coleman IM, Nguyen HM, Lin DW, Corey E, Nelson PS, Morrissey C. Cabozantinib can block growth of neuroendocrine prostate cancer patient-derived xenografts by disrupting tumor vasculature. PLoS One 2021; 16:e0245602. [PMID: 33471819 PMCID: PMC7817027 DOI: 10.1371/journal.pone.0245602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 01/04/2021] [Indexed: 12/15/2022] Open
Abstract
With the advent of potent second-line anti-androgen therapy, we and others have observed an increased incidence of androgen receptor (AR)-null small cell or neuroendocrine prostate cancer (SCNPC) in metastatic castration-resistant prostate cancer (mCRPC). Our study was designed to determine the effect of cabozantinib, a multi-targeted tyrosine kinase inhibitor that inhibits VEGFR2, MET and RET on SCNPC. Transcriptome analysis of the University of Washington rapid autopsy and SU2C mCRPC datasets revealed upregulated MET and RET expression in SCNPCs relative to adenocarcinomas. Additionally, increased MET expression correlated with attenuated AR expression and activity. In vitro treatment of SCNPC patient-derived xenograft (PDX) cells with the MET inhibitor AMG-337 had no impact on cell viability in LuCaP 93 (MET+/RET+) and LuCaP 173.1 (MET-/RET-), whereas cabozantinib decreased cell viability of LuCaP 93, but not LuCaP 173.1. Notably, MET+/RET+ LuCaP 93 and MET-/RET- LuCaP 173.1 tumor volumes were significantly decreased with cabozantinib treatment in vivo, and this activity was independent of MET or RET expression in LuCaP 173.1. Tissue analysis indicated that cabozantinib did not inhibit tumor cell proliferation (Ki67), but significantly decreased microvessel density (CD31) and increased hypoxic stress and glycolysis (HK2) in LuCaP 93 and LuCaP 173.1 tumors. RNA-Seq and gene set enrichment analysis revealed that hypoxia and glycolysis pathways were increased in cabozantinib-treated tumors relative to control tumors. Our data suggest that the most likely mechanism of cabozantinib-mediated tumor growth suppression in SCNPC PDX models is through disruption of the tumor vasculature. Thus, cabozantinib may represent a potential therapy for patients with metastatic disease in tumor phenotypes that have a significant dependence on the tumor vasculature for survival and proliferation.
Collapse
Affiliation(s)
- Mark P. Labrecque
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Lisha G. Brown
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Ilsa M. Coleman
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Holly M. Nguyen
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Daniel W. Lin
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Eva Corey
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Peter S. Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Colm Morrissey
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
13
|
Abstract
Is it true that cabozantinib should be the preferred option treating patients with bone metastases? Are there any reliable comparisons between this drug and other standard options in this subgroup? To address the issue, we performed a systematic review and metanalysis of randomized trials with cabozantinib, to assess its effectiveness, in terms of overall survival, according to the presence of bone metastases. We included (a) randomized controlled trials; (b) any solid tumors and therapeutic line; and (c) overall survival data available according to the site of disease. Cabozantinib improved overall survival both for the group with bone metastases, with risk of death decreased by 53% (hazard ratio, 0.47; 95% confidence interval, 0.26-0.87; P=0.02) and for the group without bone metastases, decreasing the risk of death by 44% (hazard ratio, 0.56; 95% confidence interval, 0.40-0.79; P=0.001) over the standard of care. The difference was not significantly different between the two groups. Despite cabozantinib can be undoubtedly listed as a good therapeutic option for cancer patients with bone metastases, it seems that its preclinical profile against bone remodeling does not translate into an actual clinical relevance, preventing from considering the presence of bone metastases as principal criterion for the choice of this drug.
Collapse
|
14
|
Rathi N, McFarland TR, Nussenzveig R, Agarwal N, Swami U. Evolving Role of Immunotherapy in Metastatic Castration Refractory Prostate Cancer. Drugs 2020; 81:191-206. [PMID: 33369720 PMCID: PMC7932934 DOI: 10.1007/s40265-020-01456-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Immunotherapies have shown remarkable success in the treatment of multiple cancer types; however, despite encouraging preclinical activity, registration trials of immunotherapy in prostate cancer have largely been unsuccessful. Sipuleucel-T remains the only approved immunotherapy for the treatment of asymptomatic or minimally symptomatic metastatic castrate-resistant prostate cancer based on modest improvement in overall survival. This immune evasion in the case of prostate cancer has been attributed to tumor-intrinsic factors, an immunosuppressive tumor microenvironment, and host factors, which ultimately make it an inert 'cold' tumor. Recently, multiple approaches have been investigated to turn prostate cancer into a 'hot' tumor. Antibodies directed against programmed cell death protein 1 have a tumor agnostic approval for a small minority of patients with microsatellite instability-high or mismatch repair-deficient metastatic prostate cancer. Herein, we present an overview of the current immunotherapy landscape in metastatic castration-resistant prostate cancer with a focus on immune checkpoint inhibitors. We describe the results of clinical trials of immune checkpoint inhibitors in patients with metastatic castration-resistant prostate cancer; either as single agents or in combination with other checkpoint inhibitors, poly (ADP-ribose) polymerase (PARP) inhibitors, tyrosine kinase inhibitors, novel hormonal therapies, chemotherapies, and radioligands. Finally, we review upcoming immunotherapies, including novel monoclonal antibodies, chimeric-antigen receptor (CAR) T cells, Bi-Specific T cell Engagers (BiTEs), therapies targeting the adenosine pathway, and other miscellaneous agents.
Collapse
Affiliation(s)
- Nityam Rathi
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive Suite 5726, Salt Lake City, UT, 84112, USA
| | - Taylor Ryan McFarland
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive Suite 5726, Salt Lake City, UT, 84112, USA
| | - Roberto Nussenzveig
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive Suite 5726, Salt Lake City, UT, 84112, USA
| | - Neeraj Agarwal
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive Suite 5726, Salt Lake City, UT, 84112, USA
| | - Umang Swami
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive Suite 5726, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
15
|
Silva Paiva R, Gomes I, Casimiro S, Fernandes I, Costa L. c-Met expression in renal cell carcinoma with bone metastases. J Bone Oncol 2020; 25:100315. [PMID: 33024658 PMCID: PMC7527574 DOI: 10.1016/j.jbo.2020.100315] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 12/12/2022] Open
Abstract
Bone is a common metastatic site in renal cell carcinoma (RCC). HGF/c-Met pathway is particularly relevant in tumors with bone metastases. c-Met/HGF pathway is involved in RCC progression, conferring poor prognosis. Several c-Met targeting therapies are currently in clinical development. c-Met expression is an important therapeutic target in RCC with bone metastases.
Hepatocyte growth factor (HGF)/c-Met pathway is implicated in embryogenesis and organ development and differentiation. Germline or somatic mutations, chromosomal rearrangements, gene amplification, and transcriptional upregulation in MET or alterations in autocrine or paracrine c-Met signalling have been associated with cancer cell proliferation and survival, including in renal cell carcinoma (RCC), and associated with disease progression. HGF/c-Met pathway has been shown to be particularly relevant in tumors with bone metastases (BMs). However, the efficacy of targeting c-Met in bone metastatic disease, including in RCC, has not been proven. Therefore, further investigation is required focusing the particular role of HGF/c-Met pathway in bone microenvironment (BME) and how to effectively target this pathway in the context of bone metastatic disease.
Collapse
Key Words
- ALK, anaplastic lymphoma kinase gene
- AR, androgen receptor
- ATP, adenosine triphosphate
- AXL, AXL Receptor Tyrosine Kinase
- BME, bone microenvironment
- BMPs, bone morphogenetic proteins
- BMs, bone metastases
- BPs, Bisphosphonates
- BTAs, Bone-targeting agents
- Bone metastases
- CCL20, chemokine (C-C motif) ligand 20
- CI, confidence interval
- CRPC, Castration Resistant Prostate Cancer
- CSC, cancer stem cells
- CTC, circulating tumor cells
- CaSR, calcium/calcium-sensing receptor
- EMA, European Medicines Agency
- EMT, epithelial-to-mesenchymal transition
- FDA, US Food and Drug Administration
- FLT-3, FMS-like tyrosine kinase 3
- GEJ, Gastroesophageal Junction
- HCC, Hepatocellular Carcinoma
- HGF, hepatocyte growth factor
- HGF/c-Met
- HIF, hypoxia-inducible factors
- HR, hazard ratio
- IGF, insulin-like growth factor
- IGF2BP3, insulin mRNA Binding Protein-3
- IL, interleukin
- IRC, independent review committees
- KIT, tyrosine-protein kinase KIT
- Kidney cancer
- M-CSF, macrophage colony-stimulating factor
- MET, MET proto-oncogene, receptor tyrosine kinase
- NSCLC, non-small cell lung carcinoma
- ORR, overall response rate
- OS, overall survival
- PDGF, platelet-derived growth factor
- PFS, progression free survival
- PTHrP, parathyroid hormone-related peptide
- RANKL, receptor activator of nuclear factor-κB ligand
- RCC, renal cell carcinoma
- RET, rearranged during transfection proto-oncogene
- ROS, proto-oncogene tyrosine-protein kinase ROS
- RTK, receptor tyrosine kinase
- SCLC, Squamous Cell Lung Cancer
- SREs, skeletal-related events
- SSE, symptomatic skeletal events
- TGF-β, transforming growth factor-β
- TIE-2, Tyrosine-Protein Kinase Receptor TIE-2
- TKI, tyrosine kinase inhibitor
- TRKB, Tropomyosin receptor kinase B
- Targeted therapy
- VEGFR, vascular endothelial growth factor receptor
- VHL, Hippel-Lindau tumor suppressor gene
- ZA, zoledronic acid
- ccRCC, clear-cell RCC
- mAb, monoclonal antibodies
- pRCC, papillary renal cell carcinoma
Collapse
Affiliation(s)
- Rita Silva Paiva
- Oncology Division, Hospital de Santa Maria, CHULN, 1649-035 Lisboa, Portugal
| | - Inês Gomes
- Instituto de Medicina Molecular - João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Sandra Casimiro
- Instituto de Medicina Molecular - João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Isabel Fernandes
- Oncology Division, Hospital de Santa Maria, CHULN, 1649-035 Lisboa, Portugal
- Instituto de Medicina Molecular - João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Luís Costa
- Oncology Division, Hospital de Santa Maria, CHULN, 1649-035 Lisboa, Portugal
- Instituto de Medicina Molecular - João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
- Corresponding author at: Oncology Division, Hospital de Santa Maria, 1649-035 Lisbon, Portugal.
| |
Collapse
|
16
|
Smith DC, Daignault-Newton S, Grivas P, Reichert ZR, Hussain M, Cooney KA, Caram M, Alva A, Jacobson J, Yablon C, Mehra R, Escara-Wilke J, Shelley G, Keller ET. Efficacy and Effect of Cabozantinib on Bone Metastases in Treatment-naive Castration-resistant Prostate Cancer. Clin Genitourin Cancer 2020; 18:332-339.e2. [PMID: 32299729 DOI: 10.1016/j.clgc.2019.10.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/21/2019] [Accepted: 10/28/2019] [Indexed: 11/27/2022]
Abstract
BACKGROUND Cabozantinib is active in advanced prostate cancer with improvement on bone scans in men on phase II trials. This trial evaluated the efficacy and changes in bone lesions in men with metastatic castration-resistant prostate cancer (mCRPC) treated with cabozantinib. PATIENTS AND METHODS Eligible patients with mCRPC involving bone underwent biopsy of a bone lesion followed by cabozantinib starting at 60 mg daily and continuing until progression or intolerable toxicity. The primary study endpoint was progression-free survival at 12 weeks. The bone lesion was rebiopsied at 6 weeks. Expression of CMET, phospho-CMET, and VEGFR2 was assayed by immunohistochemistry. Serum was obtained at baseline, and at 3, 6, and 12 weeks and assayed for bone remodeling markers. RESULTS A total of 25 patients were enrolled: 22 were evaluable, and 3 were excluded before receiving cabozantinib. At 12 weeks, 17 (77%) of 22 patients had stable disease or better. The median time on treatment was 24 weeks (range, 3-112 weeks). The overall median progression-free survival was 43.7 weeks (95% confidence interval, 23.7-97.0 weeks). Eight (36%) of 22 patients had markedly reduced uptake on bone scan. Patients with significant response on bone scan had higher bone morphogenic protein-2 levels at baseline, stable N-telopeptides levels, increased vascular endothelial growth factor receptor 2 expression, and a trend towards increased phospho-CMET while on cabozantinib compared with patients with stable disease. CONCLUSIONS Cabozantinib is active in men with mCRPC, inducing significant changes on bone scan in one-third of patients with changes in markers of bone formation and the tumor microenvironment.
Collapse
Affiliation(s)
- David C Smith
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI; University of Michigan Rogel Cancer Center, Ann Arbor, MI.
| | | | - Petros Grivas
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI; University of Michigan Rogel Cancer Center, Ann Arbor, MI
| | - Zachery R Reichert
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI; University of Michigan Rogel Cancer Center, Ann Arbor, MI
| | - Maha Hussain
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI; University of Michigan Rogel Cancer Center, Ann Arbor, MI
| | - Kathleen A Cooney
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI; University of Michigan Rogel Cancer Center, Ann Arbor, MI
| | - Megan Caram
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI; University of Michigan Rogel Cancer Center, Ann Arbor, MI
| | - Ajjai Alva
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI; University of Michigan Rogel Cancer Center, Ann Arbor, MI
| | - Jon Jacobson
- Department of Radiology, University of Michigan, Ann Arbor, MI
| | - Corrie Yablon
- Department of Radiology, University of Michigan, Ann Arbor, MI
| | - Rohit Mehra
- Department of Pathology, University of Michigan, Ann Arbor, MI
| | - June Escara-Wilke
- University of Michigan Rogel Cancer Center, Ann Arbor, MI; Department of Urology, University of Michigan, Ann Arbor, MI
| | - Greg Shelley
- University of Michigan Rogel Cancer Center, Ann Arbor, MI; Department of Urology, University of Michigan, Ann Arbor, MI
| | - Evan T Keller
- University of Michigan Rogel Cancer Center, Ann Arbor, MI; Department of Urology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
17
|
Kang J, Choi YJ, Seo BY, Jo U, Park SI, Kim YH, Park KH. A Selective FGFR inhibitor AZD4547 suppresses RANKL/M-CSF/OPG-dependent ostoclastogenesis and breast cancer growth in the metastatic bone microenvironment. Sci Rep 2019; 9:8726. [PMID: 31217507 PMCID: PMC6584658 DOI: 10.1038/s41598-019-45278-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 01/11/2019] [Indexed: 01/04/2023] Open
Abstract
Aberrant activation of fibroblast growth factor receptor (FGFR) signalling contributes to progression and metastasis of many types of cancers including breast cancer. Accordingly, FGFR targeted tyrosine kinase inhibitors (TKIs) are currently under development. However, the efficacy of FGFR TKIs in the bone microenvironment where breast cancer cells most frequently metastasize and also where FGFR is biologically active, has not been clearly investigated. We investigated the FGFR-mediated interactions among cancer and the bone microenvironment stromal cells (osteoblasts and osteoclasts), and also the effects of FGFR inhibition in bone metastasis. We showed that addition of culture supernatant from the MDA-MB-134-VI FGFR-amplified breast cancer cells-activated FGFR siganalling in osteoblasts, including increased expression of RANKL, M-CSF, and osteoprotegerin (OPG). Further in vitro analyses showed that AZD4547, an FGFR TKI currently in clinical trials for breast cancer, decreased RANKL and M-CSF, and subsequently RANKL and M-CSF-dependent osteoclastogenesis of murine bone marrow monocytes. Moreover, AZD4547 suppressed osteoclastogenesis and tumor-induced osteolysis in an orthotopic breast cancer bone metastasis mouse model using FGFR non-amplified MDA-MB-231 cells. Collectively, our results support that FGFR inhibitors inhibit the bone microenvironment stromal cells including osteoblasts and osteoclasts, and effectively suppress both tumor and stromal compartments of bone metastasis.
Collapse
Affiliation(s)
- Jinho Kang
- The BK21 Plus Program, Korea University College of Medicine, Seongbuk-Gu, Seoul, Republic of Korea.,Division of Oncology/Hematology, Department of Internal Medicine, Korea University College of Medicine, Seongbuk-Gu, Seoul, Republic of Korea
| | - Yoon Ji Choi
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University College of Medicine, Seongbuk-Gu, Seoul, Republic of Korea
| | - Bo Yeon Seo
- The BK21 Plus Program, Korea University College of Medicine, Seongbuk-Gu, Seoul, Republic of Korea.,Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seongbuk-Gu, Seoul, Republic of Korea
| | - Ukhyun Jo
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University College of Medicine, Seongbuk-Gu, Seoul, Republic of Korea
| | - Serk In Park
- The BK21 Plus Program, Korea University College of Medicine, Seongbuk-Gu, Seoul, Republic of Korea.,Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seongbuk-Gu, Seoul, Republic of Korea.,Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Yeul Hong Kim
- The BK21 Plus Program, Korea University College of Medicine, Seongbuk-Gu, Seoul, Republic of Korea.,Division of Oncology/Hematology, Department of Internal Medicine, Korea University College of Medicine, Seongbuk-Gu, Seoul, Republic of Korea
| | - Kyong Hwa Park
- The BK21 Plus Program, Korea University College of Medicine, Seongbuk-Gu, Seoul, Republic of Korea. .,Division of Oncology/Hematology, Department of Internal Medicine, Korea University College of Medicine, Seongbuk-Gu, Seoul, Republic of Korea.
| |
Collapse
|
18
|
Lam HM, Nguyen HM, Labrecque MP, Brown LG, Coleman IM, Gulati R, Lakely B, Sondheim D, Chatterjee P, Marck BT, Matsumoto AM, Mostaghel EA, Schweizer MT, Nelson PS, Corey E. Durable Response of Enzalutamide-resistant Prostate Cancer to Supraphysiological Testosterone Is Associated with a Multifaceted Growth Suppression and Impaired DNA Damage Response Transcriptomic Program in Patient-derived Xenografts. Eur Urol 2019; 77:144-155. [PMID: 31227306 DOI: 10.1016/j.eururo.2019.05.042] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 05/30/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Androgen deprivation therapy improves the survival of castration-resistant prostate cancer (CRPC) patients, yet ultimately fails with debilitating side effects. Supraphysiological testosterone (SPT)-based therapy produces clinical responses with improved quality of life in a subset of patients. Currently, no information defines a durable response to SPT. OBJECTIVE To identify key molecular phenotypes underlying SPT response to improve patient selection and guide combination treatment to achieve a durable response. DESIGN, SETTING, AND PARTICIPANTS A patient-derived xenograft (PDX) preclinical trial was performed with 13 CRPC PDXs to identify molecular features associated with SPT response. Comprehensive intratumoral androgen, tumor growth, and integrated transcriptomic and protein analyses were performed in three PDXs resistant to the newer androgen receptor (AR) pathway inhibitor enzalutamide (ENZ) to define SPT response and resistance. INTERVENTION Testosterone cypionate. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS SPT efficacy was evaluated by PDX growth, prostate-specific antigen (PSA) change, and survival. Intratumoral androgens were analyzed using mass spectrometry. Global transcriptome analysis was performed using RNA sequencing, and confirmed by quantitative real-time polymerase chain reaction and immunohistochemistry. Log-rank and Mann-Whitney tests were used for survival and molecular analyses, respectively. RESULTS AND LIMITATIONS A durable SPT responder was identified, presenting robust repressions of ARv7 and E2F transcriptional outputs, and a DNA damage response (DDR) transcriptomic program that were altogether restored upon SPT resistance in the transient responder. ENZ rechallenge of SPT-relapsed PDXs resulted in PSA decreases but tumor progression. CONCLUSIONS SPT produces a durable response in AR-pathway inhibitor ENZ CRPC that is associated with sustained suppression of ARv7 and E2F transcriptional outputs, and the DDR transcriptome, highlighting the potential of combination treatments that maintain suppression of these programs to drive a durable response to SPT. PATIENT SUMMARY Patients with ENZ-resistant prostate cancer have very limited treatment options. Supraphysiological testosterone presents a prominent option for improved quality of life and a potential durable response in patients with sustained suppression on ARv7/E2F transcriptional outputs and DNA repair program.
Collapse
Affiliation(s)
- Hung-Ming Lam
- Department of Urology, University of Washington School of Medicine, Seattle, WA, USA; Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Holly M Nguyen
- Department of Urology, University of Washington School of Medicine, Seattle, WA, USA
| | - Mark P Labrecque
- Department of Urology, University of Washington School of Medicine, Seattle, WA, USA
| | - Lisha G Brown
- Department of Urology, University of Washington School of Medicine, Seattle, WA, USA
| | - Ilsa M Coleman
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Roman Gulati
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Bryce Lakely
- Department of Urology, University of Washington School of Medicine, Seattle, WA, USA
| | - Daniel Sondheim
- Department of Urology, University of Washington School of Medicine, Seattle, WA, USA
| | - Payel Chatterjee
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Brett T Marck
- Geriatric Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Alvin M Matsumoto
- Geriatric Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA; Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA, USA; Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA, USA
| | - Elahe A Mostaghel
- Geriatric Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA; Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA, USA
| | - Michael T Schweizer
- Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA, USA; Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Peter S Nelson
- Department of Urology, University of Washington School of Medicine, Seattle, WA, USA; Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA, USA
| | - Eva Corey
- Department of Urology, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
19
|
Basch EM, Scholz M, de Bono JS, Vogelzang N, de Souza P, Marx G, Vaishampayan U, George S, Schwarz JK, Antonarakis ES, O'Sullivan JM, Kalebasty AR, Chi KN, Dreicer R, Hutson TE, Dueck AC, Bennett AV, Dayan E, Mangeshkar M, Holland J, Weitzman AL, Scher HI. Cabozantinib Versus Mitoxantrone-prednisone in Symptomatic Metastatic Castration-resistant Prostate Cancer: A Randomized Phase 3 Trial with a Primary Pain Endpoint. Eur Urol 2019; 75:929-937. [PMID: 30528222 PMCID: PMC6876845 DOI: 10.1016/j.eururo.2018.11.033] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 11/14/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND Bone metastases in patients with metastatic castration-resistant prostate cancer (mCRPC) are associated with debilitating pain and functional compromise. OBJECTIVE To compare pain palliation as the primary endpoint for cabozantinib versus mitoxantrone-prednisone in men with mCRPC and symptomatic bone metastases using patient-reported outcome measures. DESIGN, SETTING, AND PARTICIPANTS A randomized, double-blind phase 3 trial (COMET-2; NCT01522443) in men with mCRPC and narcotic-dependent pain from bone metastases who had progressed after treatment with docetaxel and either abiraterone or enzalutamide. INTERVENTION Cabozantinib 60mg once daily orally versus mitoxantrone 12mg/m2 every 3wk plus prednisone 5mg twice daily orally. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The primary endpoint was pain response at week 6 confirmed at week 12 (≥30% decrease from baseline in patient-reported average daily worst pain score via the Brief Pain Inventory without increased narcotic use). The planned sample size was 246 to achieve ≥90% power. RESULTS AND LIMITATIONS Enrollment was terminated early because cabozantinib did not demonstrate a survival benefit in the companion COMET-1 trial. At study closure, 119 participants were randomized (cabozantinib: N=61; mitoxantrone-prednisone: N=58). Complete pain and narcotic use data were available at baseline, week 6, and week 12 for 73/106 (69%) patients. There was no significant difference in the pain response with cabozantinib versus mitoxantrone-prednisone: the proportions of responders were 15% versus 17%, a -2% difference (95% confidence interval: -16% to 11%, p=0.8). Barriers to accrual included pretreatment requirements for a washout period of prior anticancer therapy and a narcotic optimization period to maximize analgesic dosing. CONCLUSIONS Cabozantinib treatment did not demonstrate better pain palliation than mitoxantrone-prednisone in heavily pretreated patients with mCRPC and symptomatic bone metastases. Future pain-palliation trials should incorporate briefer timelines from enrollment to treatment initiation. PATIENT SUMMARY Cabozantinib was not better than mitoxantrone-prednisone for pain relief in patients with castration-resistant prostate cancer and debilitating pain from bone metastases.
Collapse
Affiliation(s)
- Ethan M Basch
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.
| | - Mark Scholz
- Prostate Oncology Specialists, Marina del Rey, CA, USA
| | - Johann S de Bono
- Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, England, UK
| | | | - Paul de Souza
- Western Sydney University School of Medicine, Sydney, Australia
| | - Gavin Marx
- Sydney Medical School, University of Sydney and Sydney Adventist Hospital, Wahroonga, Australia
| | | | - Saby George
- Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | | | | | | | - Kim N Chi
- British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Robert Dreicer
- Emily Couric Clinical Cancer Center, University of Virginia, Charlottesville, VA, USA
| | | | - Amylou C Dueck
- Department of Health Sciences Research, Mayo Clinic, Scottsdale, AZ, USA
| | - Antonia V Bennett
- Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Erica Dayan
- Department of Medicine, Genitourinary Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | | | - Howard I Scher
- Department of Medicine, Genitourinary Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
20
|
D'Oronzo S, Coleman R, Brown J, Silvestris F. Metastatic bone disease: Pathogenesis and therapeutic options: Up-date on bone metastasis management. J Bone Oncol 2019; 15:004-4. [PMID: 30937279 PMCID: PMC6429006 DOI: 10.1016/j.jbo.2018.10.004] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/22/2018] [Accepted: 10/28/2018] [Indexed: 12/17/2022] Open
Abstract
Bone metastases (BM) are a common complication of cancer, whose management often requires a multidisciplinary approach. Despite the recent therapeutic advances, patients with BM may still experience skeletal-related events and symptomatic skeletal events, with detrimental impact on quality of life and survival. A deeper knowledge of the mechanisms underlying the onset of lytic and sclerotic BM has been acquired in the last decades, leading to the development of bone-targeting agents (BTA), mainly represented by anti-resorptive drugs and bone-seeking radiopharmaceuticals. Recent pre-clinical and clinical studies have showed promising effects of novel agents, whose safety and efficacy need to be confirmed by prospective clinical trials. Among BTA, adjuvant bisphosphonates have also been shown to reduce the risk of BM in selected breast cancer patients, but failed to reduce the incidence of BM from lung and prostate cancer. Moreover, adjuvant denosumab did not improve BM free survival in patients with breast cancer, suggesting the need for further investigation to clarify BTA role in early-stage malignancies. The aim of this review is to describe BM pathogenesis and current treatment options in different clinical settings, as well as to explore the mechanism of action of novel potential therapeutic agents for which further investigation is needed.
Collapse
Key Words
- ActRIIA, activin-A type IIA receptor
- BC, breast cancer
- BM, bone metastases
- BMD, bone mineral density
- BMPs, bone morphogenetic proteins
- BMSC, bone marrow stromal cells
- BPs, bisphosphonates
- BTA, bone targeting agents
- BTM, bone turnover markers
- Bone metastases
- Bone targeting agents
- CCR, chemokine-receptor
- CRPC, castration-resistant PC
- CXCL-12, C–X–C motif chemokine-ligand-12
- CXCR-4, chemokine-receptor-4
- DFS, disease-free survival
- DKK1, dickkopf1
- EBC, early BC
- ECM, extracellular matrix
- ET-1, endothelin-1
- FDA, food and drug administration
- FGF, fibroblast growth factor
- GAS6, growth-arrest specific-6
- GFs, growth factors
- GnRH, gonadotropin-releasing hormone
- HER-2, human epidermal growth factor receptor 2
- HR, hormone receptor
- IL, interleukin
- LC, lung cancer
- MAPK, mitogen-activated protein kinase
- MCSF, macrophage colony-stimulating factor
- MCSFR, MCSF receptor
- MIP-1α, macrophage inflammatory protein-1 alpha
- MM, multiple myeloma
- MPC, malignant plasma cells
- N-BPs, nitrogen-containing BPs
- NF-κB, nuclear factor-κB
- ONJ, osteonecrosis of the jaw
- OS, overall survival
- Osteotropic tumors
- PC, prostate cancer
- PDGF, platelet-derived growth factor
- PFS, progression-free survival
- PIs, proteasome inhibitors
- PSA, prostate specific antigen
- PTH, parathyroid hormone
- PTH-rP, PTH related protein
- QoL, quality of life
- RANK-L, receptor activator of NF-κB ligand
- RT, radiation therapy
- SREs, skeletal-related events
- SSEs, symptomatic skeletal events
- Skeletal related events
- TGF-β, transforming growth factor β
- TK, tyrosine kinase
- TKIs, TK inhibitors
- TNF, tumornecrosis factor
- VEGF, vascular endothelial growth factor
- VEGFR, VEGF receptor
- mTOR, mammalian target of rapamycin
- non-N-BPs, non-nitrogen containing BPs
- v-ATPase, vacuolar-type H+ ATPase
Collapse
Affiliation(s)
- Stella D'Oronzo
- Medical Oncology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, P.za Giulio Cesare, 11, 70124 Bari, Italy
| | - Robert Coleman
- Academic Unit of Clinical Oncology, Weston Park Hospital, University of Sheffield, Whitham Rd, Sheffield S10 2SJ, England, UK
| | - Janet Brown
- Academic Unit of Clinical Oncology, Weston Park Hospital, University of Sheffield, Whitham Rd, Sheffield S10 2SJ, England, UK
| | - Francesco Silvestris
- Medical Oncology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, P.za Giulio Cesare, 11, 70124 Bari, Italy
| |
Collapse
|
21
|
Puccini A, Marín-Ramos NI, Bergamo F, Schirripa M, Lonardi S, Lenz HJ, Loupakis F, Battaglin F. Safety and Tolerability of c-MET Inhibitors in Cancer. Drug Saf 2019; 42:211-233. [PMID: 30649748 PMCID: PMC7491978 DOI: 10.1007/s40264-018-0780-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The role of aberrant hepatocyte growth factor receptor (c-MET, also known as tyrosine-protein kinase MET)/hepatocyte growth factor (HGF) signaling in cancer progression and invasion has been extensively studied. c-MET inhibitors have shown promising pre-clinical and early phase clinical trial anti-tumor activity in several tumor types, although results of most phase III trials with these agents have been negative. To date, two small molecule c-MET inhibitors, cabozantinib and crizotinib, have been approved by regulatory authorities for the treatment of selected cancer types, but several novel c-MET inhibitors (either monoclonal antibodies or small molecule c-MET tyrosine kinase inhibitors) and treatment combinations are currently under study in different settings. Here we provide an overview of the mechanism of action and rationale of c-MET inhibition in cancer, the efficacy of approved agents, and novel promising c-MET-inhibitors and novel targeted combination strategies under development in different cancer types, with a focus on the safety profile and tolerability of these compounds.
Collapse
Affiliation(s)
- Alberto Puccini
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Suite 5410, Los Angeles, CA, 90033, USA
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Nagore I Marín-Ramos
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Francesca Bergamo
- Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Marta Schirripa
- Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Sara Lonardi
- Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Suite 5410, Los Angeles, CA, 90033, USA
| | - Fotios Loupakis
- Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Francesca Battaglin
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Suite 5410, Los Angeles, CA, 90033, USA.
- Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy.
| |
Collapse
|
22
|
Scott AJ, Arcaroli JJ, Bagby SM, Yahn R, Huber KM, Serkova NJ, Nguyen A, Kim J, Thorburn A, Vogel J, Quackenbush KS, Capasso A, Schreiber A, Blatchford P, Klauck PJ, Pitts TM, Eckhardt SG, Messersmith WA. Cabozantinib Exhibits Potent Antitumor Activity in Colorectal Cancer Patient-Derived Tumor Xenograft Models via Autophagy and Signaling Mechanisms. Mol Cancer Ther 2018; 17:2112-2122. [PMID: 30026382 DOI: 10.1158/1535-7163.mct-17-0131] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 12/01/2017] [Accepted: 07/09/2018] [Indexed: 12/13/2022]
Abstract
Antiangiogenic therapy used in treatment of metastatic colorectal cancer (mCRC) inevitably succumbs to treatment resistance. Upregulation of MET may play an essential role to acquired anti-VEGF resistance. We previously reported that cabozantinib (XL184), an inhibitor of receptor tyrosine kinases (RTK) including MET, AXL, and VEGFR2, had potent antitumor effects in mCRC patient-derived tumor explant models. In this study, we examined the mechanisms of cabozantinib sensitivity, using regorafenib as a control. The tumor growth inhibition index (TGII) was used to compare treatment effects of cabozantinib 30 mg/kg daily versus regorafenib 10 mg/kg daily for a maximum of 28 days in 10 PDX mouse models. In vivo angiogenesis and glucose uptake were assessed using dynamic contrast-enhanced (DCE)-MRI and [18F]-FDG-PET imaging, respectively. RNA-Seq, RTK assay, and immunoblotting analysis were used to evaluate gene pathway regulation in vivo and in vitro Analysis of TGII demonstrated significant antitumor effects with cabozantinib compared with regorafenib (average TGII 3.202 vs. 48.48, respectively; P = 0.007). Cabozantinib significantly reduced vascularity and glucose uptake compared with baseline. Gene pathway analysis showed that cabozantinib significantly decreased protein activity involved in glycolysis and upregulated proteins involved in autophagy compared with control, whereas regorafenib did not. The combination of two separate antiautophagy agents, SBI-0206965 and chloroquine, plus cabozantinib increased apoptosis in vitro Cabozantinib demonstrated significant antitumor activity, reduction in tumor vascularity, increased autophagy, and altered cell metabolism compared with regorafenib. Our findings support further evaluation of cabozantinib and combinational approaches targeting autophagy in colorectal cancer. Mol Cancer Ther; 17(10); 2112-22. ©2018 AACR.
Collapse
Affiliation(s)
- Aaron J Scott
- Division of Hematology and Oncology, Banner University of Arizona Cancer Center, Tucson, Arizona.
| | - John J Arcaroli
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus and University of Colorado Cancer Center, Aurora, Colorado
| | - Stacey M Bagby
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus and University of Colorado Cancer Center, Aurora, Colorado
| | - Rachel Yahn
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus and University of Colorado Cancer Center, Aurora, Colorado
| | - Kendra M Huber
- Department of Anesthesia, University of Colorado Anschutz Medical Campus and University of Colorado Cancer Center, Aurora, Colorado
| | - Natalie J Serkova
- Department of Anesthesia, University of Colorado Anschutz Medical Campus and University of Colorado Cancer Center, Aurora, Colorado
| | - Anna Nguyen
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus and University of Colorado Cancer Center, Aurora, Colorado
| | - Jihye Kim
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus and University of Colorado Cancer Center, Aurora, Colorado
| | - Andrew Thorburn
- Department of Pharmacology, University of Colorado Anschutz Medical Campus and University of Colorado Cancer Center, Aurora, Colorado
| | - Jon Vogel
- Department of Surgery, University of Colorado Anschutz Medical Campus and University of Colorado Cancer Center, Aurora, Colorado
| | - Kevin S Quackenbush
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus and University of Colorado Cancer Center, Aurora, Colorado
| | - Anna Capasso
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus and University of Colorado Cancer Center, Aurora, Colorado
| | - Anna Schreiber
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus and University of Colorado Cancer Center, Aurora, Colorado
| | - Patrick Blatchford
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus and University of Colorado Cancer Center, Aurora, Colorado
| | - Peter J Klauck
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus and University of Colorado Cancer Center, Aurora, Colorado
| | - Todd M Pitts
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus and University of Colorado Cancer Center, Aurora, Colorado
| | - S Gail Eckhardt
- Division of Medical Oncology, The University of Texas at Austin, Austin, Texas
| | - Wells A Messersmith
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus and University of Colorado Cancer Center, Aurora, Colorado
| |
Collapse
|
23
|
Ren LJ, Wu HJ, Sun LH, Xu X, Mo LY, Zhang L, Zhang JY, Wu CY. A sensitive LC-MS/MS method for simultaneous determination of cabozantinib and its metabolite cabozantinib N
-oxide in rat plasma and its application in a pharmacokinetic study. Biomed Chromatogr 2018; 32:e4227. [DOI: 10.1002/bmc.4227] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 02/22/2018] [Accepted: 02/23/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Lian-jie Ren
- Department of Pharmaceutics, School of Pharmacy; China Pharmaceutical University; Nanjing China
- Center for Drug Evaluation; China Food and Drug Administration; Beijing China
| | - Hua-jing Wu
- Department of Pharmaceutical Analysis; China Pharmaceutical University; Nanjing China
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research; Shanghai Institute of Materia Medica, Chinese Academy of Sciences; Shanghai China
| | - Li-han Sun
- Department of Pharmaceutical Analysis; China Pharmaceutical University; Nanjing China
| | - Xue Xu
- Department of Pharmaceutical Analysis; China Pharmaceutical University; Nanjing China
| | - Li-ying Mo
- Department of Pharmaceutics of Traditional Chinese Medicine; China Pharmaceutical University; Nanjing China
| | - Lei Zhang
- Department of Chemical Drug Division; Shandong Institute for Food and Drug Control; Shandong China
| | - Jun-ying Zhang
- Department of Pharmaceutics of Traditional Chinese Medicine; China Pharmaceutical University; Nanjing China
| | - Chun-yong Wu
- Department of Pharmaceutical Analysis; China Pharmaceutical University; Nanjing China
| |
Collapse
|
24
|
Sousa S, Clézardin P. Bone-Targeted Therapies in Cancer-Induced Bone Disease. Calcif Tissue Int 2018; 102:227-250. [PMID: 29079995 DOI: 10.1007/s00223-017-0353-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 10/19/2017] [Indexed: 01/14/2023]
Abstract
Cancer-induced bone disease is a major source of morbidity and mortality in cancer patients. Thus, effective bone-targeted therapies are essential to improve disease-free, overall survival and quality of life of cancer patients with bone metastases. Depending of the cancer-type, bone metastases mainly involve the modulation of osteoclast and/or osteoblast activity by tumour cells. To inhibit metastatic bone disease effectively, it is imperative to understand its underlying mechanisms and identify the target cells for therapy. If the aim is to prevent bone metastasis, it is essential to target not only bone metastatic features in the tumour cells, but also tumour-nurturing bone microenvironment properties. The currently available bone-targeted agents mainly affect osteoclasts, inhibiting bone resorption (e.g. bisphosphonates, denosumab). Some agents targeting osteoblasts begin to emerge which target osteoblasts (e.g. romosozumab), activating bone formation. Moreover, certain drugs initially thought to target only osteoclasts are now known to have a dual action (activating osteoblasts and inhibiting osteoclasts, e.g. proteasome inhibitors). This review will focus on the evolution of bone-targeted therapies for the treatment of cancer-induced bone disease, summarizing preclinical and clinical findings obtained with anti-resorptive and bone anabolic therapies.
Collapse
Affiliation(s)
- Sofia Sousa
- National Institute of Health and Medical Research (INSERM), UMR 1033, 69372, Lyon, France.
- Faculty of Medicine Laennec, University of Lyon-1, 69372, Villeurbanne, France.
| | - Philippe Clézardin
- National Institute of Health and Medical Research (INSERM), UMR 1033, 69372, Lyon, France
- Faculty of Medicine Laennec, University of Lyon-1, 69372, Villeurbanne, France
- European Cancer and Bone Metastasis Laboratory, Department of Bone Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, UK
| |
Collapse
|
25
|
Escudier B, Powles T, Motzer RJ, Olencki T, Arén Frontera O, Oudard S, Rolland F, Tomczak P, Castellano D, Appleman LJ, Drabkin H, Vaena D, Milwee S, Youkstetter J, Lougheed JC, Bracarda S, Choueiri TK. Cabozantinib, a New Standard of Care for Patients With Advanced Renal Cell Carcinoma and Bone Metastases? Subgroup Analysis of the METEOR Trial. J Clin Oncol 2018; 36:765-772. [PMID: 29309249 DOI: 10.1200/jco.2017.74.7352] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Purpose Cabozantinib, an inhibitor of tyrosine kinases including MET, vascular endothelial growth factor receptors, and AXL, increased progression-free survival (PFS), overall survival (OS), and objective response rate (ORR) in patients with advanced renal cell carcinoma (RCC) after previous vascular endothelial growth factor receptor-targeted therapy in the phase III METEOR trial. Because bone metastases are associated with increased morbidity in patients with RCC, bone-related outcomes were analyzed in METEOR. Patients and Methods Six hundred fifty-eight patients were randomly assigned 1:1 to receive 60 mg cabozantinib or 10 mg everolimus. Prespecified subgroup analyses of PFS, OS, and ORR were conducted in patients grouped by baseline bone metastases status per independent radiology committee (IRC). Additional end points included bone scan response per IRC, skeletal-related events, and changes in bone biomarkers. Results For patients with bone metastases at baseline (cabozantinib [n = 77]; everolimus [n = 65]), median PFS was 7.4 months for cabozantinib versus 2.7 months for everolimus (hazard ratio, 0.33 [95% CI, 0.21 to 0.51]). Median OS was also longer with cabozantinib (20.1 months v 12.1 months; hazard ratio, 0.54 [95% CI, 0.34 to 0.84]), and ORR per IRC was higher (17% v 0%). The rate of skeletal-related events was 23% with cabozantinib and 29% with everolimus, and bone scan response per IRC was 20% versus 10%, respectively. PFS, OS, and ORR were also improved with cabozantinib in patients without bone metastases. Changes in bone biomarkers were greater with cabozantinib than with everolimus. The overall safety profiles of cabozantinib and everolimus in patients with bone metastases were consistent with those observed in patients without bone metastases. Conclusion Cabozantinib treatment was associated with improved PFS, OS, and ORR when compared with everolimus treatment in patients with advanced RCC and bone metastases and represents a good treatment option for these patients.
Collapse
Affiliation(s)
- Bernard Escudier
- Bernard Escudier, Institut Gustave Roussy, Villejuif; Stephane Oudard, Hôpital Européen Georges Pompidou, Paris; Frederic Rolland, Centre René Gauducheau Centre de Lutte Contre Le Cancer Nantes, Saint-Herblain, France; Thomas Powles, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University, Columbus, OH; Osvaldo Arén Frontera, Centro Internacional de Estudios Clinicos, Santiago, Chile; Piotr Tomczak, Szpital Kliniczny Przemienienia Panskiego Uniwersytetu Medycznego, Poznań, Poland; Daniel Castellano, Hospital Universitario 12 de Octubre, Madrid, Spain; Leonard J Appleman, University of Pittsburgh Medical Center, Pittsburgh, PA; Harry Drabkin, Medical University of South Carolina, Charleston, SC; Daniel Vaena, University of Iowa Hospitals and Clinics, Iowa City, IA; Steven Milwee, Jillian Youkstetter, and Julie C. Lougheed, Exelixis, Inc., San Francisco, CA; Sergio Bracarda, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Thomas Powles
- Bernard Escudier, Institut Gustave Roussy, Villejuif; Stephane Oudard, Hôpital Européen Georges Pompidou, Paris; Frederic Rolland, Centre René Gauducheau Centre de Lutte Contre Le Cancer Nantes, Saint-Herblain, France; Thomas Powles, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University, Columbus, OH; Osvaldo Arén Frontera, Centro Internacional de Estudios Clinicos, Santiago, Chile; Piotr Tomczak, Szpital Kliniczny Przemienienia Panskiego Uniwersytetu Medycznego, Poznań, Poland; Daniel Castellano, Hospital Universitario 12 de Octubre, Madrid, Spain; Leonard J Appleman, University of Pittsburgh Medical Center, Pittsburgh, PA; Harry Drabkin, Medical University of South Carolina, Charleston, SC; Daniel Vaena, University of Iowa Hospitals and Clinics, Iowa City, IA; Steven Milwee, Jillian Youkstetter, and Julie C. Lougheed, Exelixis, Inc., San Francisco, CA; Sergio Bracarda, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Robert J Motzer
- Bernard Escudier, Institut Gustave Roussy, Villejuif; Stephane Oudard, Hôpital Européen Georges Pompidou, Paris; Frederic Rolland, Centre René Gauducheau Centre de Lutte Contre Le Cancer Nantes, Saint-Herblain, France; Thomas Powles, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University, Columbus, OH; Osvaldo Arén Frontera, Centro Internacional de Estudios Clinicos, Santiago, Chile; Piotr Tomczak, Szpital Kliniczny Przemienienia Panskiego Uniwersytetu Medycznego, Poznań, Poland; Daniel Castellano, Hospital Universitario 12 de Octubre, Madrid, Spain; Leonard J Appleman, University of Pittsburgh Medical Center, Pittsburgh, PA; Harry Drabkin, Medical University of South Carolina, Charleston, SC; Daniel Vaena, University of Iowa Hospitals and Clinics, Iowa City, IA; Steven Milwee, Jillian Youkstetter, and Julie C. Lougheed, Exelixis, Inc., San Francisco, CA; Sergio Bracarda, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Thomas Olencki
- Bernard Escudier, Institut Gustave Roussy, Villejuif; Stephane Oudard, Hôpital Européen Georges Pompidou, Paris; Frederic Rolland, Centre René Gauducheau Centre de Lutte Contre Le Cancer Nantes, Saint-Herblain, France; Thomas Powles, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University, Columbus, OH; Osvaldo Arén Frontera, Centro Internacional de Estudios Clinicos, Santiago, Chile; Piotr Tomczak, Szpital Kliniczny Przemienienia Panskiego Uniwersytetu Medycznego, Poznań, Poland; Daniel Castellano, Hospital Universitario 12 de Octubre, Madrid, Spain; Leonard J Appleman, University of Pittsburgh Medical Center, Pittsburgh, PA; Harry Drabkin, Medical University of South Carolina, Charleston, SC; Daniel Vaena, University of Iowa Hospitals and Clinics, Iowa City, IA; Steven Milwee, Jillian Youkstetter, and Julie C. Lougheed, Exelixis, Inc., San Francisco, CA; Sergio Bracarda, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Osvaldo Arén Frontera
- Bernard Escudier, Institut Gustave Roussy, Villejuif; Stephane Oudard, Hôpital Européen Georges Pompidou, Paris; Frederic Rolland, Centre René Gauducheau Centre de Lutte Contre Le Cancer Nantes, Saint-Herblain, France; Thomas Powles, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University, Columbus, OH; Osvaldo Arén Frontera, Centro Internacional de Estudios Clinicos, Santiago, Chile; Piotr Tomczak, Szpital Kliniczny Przemienienia Panskiego Uniwersytetu Medycznego, Poznań, Poland; Daniel Castellano, Hospital Universitario 12 de Octubre, Madrid, Spain; Leonard J Appleman, University of Pittsburgh Medical Center, Pittsburgh, PA; Harry Drabkin, Medical University of South Carolina, Charleston, SC; Daniel Vaena, University of Iowa Hospitals and Clinics, Iowa City, IA; Steven Milwee, Jillian Youkstetter, and Julie C. Lougheed, Exelixis, Inc., San Francisco, CA; Sergio Bracarda, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Stephane Oudard
- Bernard Escudier, Institut Gustave Roussy, Villejuif; Stephane Oudard, Hôpital Européen Georges Pompidou, Paris; Frederic Rolland, Centre René Gauducheau Centre de Lutte Contre Le Cancer Nantes, Saint-Herblain, France; Thomas Powles, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University, Columbus, OH; Osvaldo Arén Frontera, Centro Internacional de Estudios Clinicos, Santiago, Chile; Piotr Tomczak, Szpital Kliniczny Przemienienia Panskiego Uniwersytetu Medycznego, Poznań, Poland; Daniel Castellano, Hospital Universitario 12 de Octubre, Madrid, Spain; Leonard J Appleman, University of Pittsburgh Medical Center, Pittsburgh, PA; Harry Drabkin, Medical University of South Carolina, Charleston, SC; Daniel Vaena, University of Iowa Hospitals and Clinics, Iowa City, IA; Steven Milwee, Jillian Youkstetter, and Julie C. Lougheed, Exelixis, Inc., San Francisco, CA; Sergio Bracarda, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Frederic Rolland
- Bernard Escudier, Institut Gustave Roussy, Villejuif; Stephane Oudard, Hôpital Européen Georges Pompidou, Paris; Frederic Rolland, Centre René Gauducheau Centre de Lutte Contre Le Cancer Nantes, Saint-Herblain, France; Thomas Powles, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University, Columbus, OH; Osvaldo Arén Frontera, Centro Internacional de Estudios Clinicos, Santiago, Chile; Piotr Tomczak, Szpital Kliniczny Przemienienia Panskiego Uniwersytetu Medycznego, Poznań, Poland; Daniel Castellano, Hospital Universitario 12 de Octubre, Madrid, Spain; Leonard J Appleman, University of Pittsburgh Medical Center, Pittsburgh, PA; Harry Drabkin, Medical University of South Carolina, Charleston, SC; Daniel Vaena, University of Iowa Hospitals and Clinics, Iowa City, IA; Steven Milwee, Jillian Youkstetter, and Julie C. Lougheed, Exelixis, Inc., San Francisco, CA; Sergio Bracarda, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Piotr Tomczak
- Bernard Escudier, Institut Gustave Roussy, Villejuif; Stephane Oudard, Hôpital Européen Georges Pompidou, Paris; Frederic Rolland, Centre René Gauducheau Centre de Lutte Contre Le Cancer Nantes, Saint-Herblain, France; Thomas Powles, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University, Columbus, OH; Osvaldo Arén Frontera, Centro Internacional de Estudios Clinicos, Santiago, Chile; Piotr Tomczak, Szpital Kliniczny Przemienienia Panskiego Uniwersytetu Medycznego, Poznań, Poland; Daniel Castellano, Hospital Universitario 12 de Octubre, Madrid, Spain; Leonard J Appleman, University of Pittsburgh Medical Center, Pittsburgh, PA; Harry Drabkin, Medical University of South Carolina, Charleston, SC; Daniel Vaena, University of Iowa Hospitals and Clinics, Iowa City, IA; Steven Milwee, Jillian Youkstetter, and Julie C. Lougheed, Exelixis, Inc., San Francisco, CA; Sergio Bracarda, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Daniel Castellano
- Bernard Escudier, Institut Gustave Roussy, Villejuif; Stephane Oudard, Hôpital Européen Georges Pompidou, Paris; Frederic Rolland, Centre René Gauducheau Centre de Lutte Contre Le Cancer Nantes, Saint-Herblain, France; Thomas Powles, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University, Columbus, OH; Osvaldo Arén Frontera, Centro Internacional de Estudios Clinicos, Santiago, Chile; Piotr Tomczak, Szpital Kliniczny Przemienienia Panskiego Uniwersytetu Medycznego, Poznań, Poland; Daniel Castellano, Hospital Universitario 12 de Octubre, Madrid, Spain; Leonard J Appleman, University of Pittsburgh Medical Center, Pittsburgh, PA; Harry Drabkin, Medical University of South Carolina, Charleston, SC; Daniel Vaena, University of Iowa Hospitals and Clinics, Iowa City, IA; Steven Milwee, Jillian Youkstetter, and Julie C. Lougheed, Exelixis, Inc., San Francisco, CA; Sergio Bracarda, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Leonard J Appleman
- Bernard Escudier, Institut Gustave Roussy, Villejuif; Stephane Oudard, Hôpital Européen Georges Pompidou, Paris; Frederic Rolland, Centre René Gauducheau Centre de Lutte Contre Le Cancer Nantes, Saint-Herblain, France; Thomas Powles, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University, Columbus, OH; Osvaldo Arén Frontera, Centro Internacional de Estudios Clinicos, Santiago, Chile; Piotr Tomczak, Szpital Kliniczny Przemienienia Panskiego Uniwersytetu Medycznego, Poznań, Poland; Daniel Castellano, Hospital Universitario 12 de Octubre, Madrid, Spain; Leonard J Appleman, University of Pittsburgh Medical Center, Pittsburgh, PA; Harry Drabkin, Medical University of South Carolina, Charleston, SC; Daniel Vaena, University of Iowa Hospitals and Clinics, Iowa City, IA; Steven Milwee, Jillian Youkstetter, and Julie C. Lougheed, Exelixis, Inc., San Francisco, CA; Sergio Bracarda, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Harry Drabkin
- Bernard Escudier, Institut Gustave Roussy, Villejuif; Stephane Oudard, Hôpital Européen Georges Pompidou, Paris; Frederic Rolland, Centre René Gauducheau Centre de Lutte Contre Le Cancer Nantes, Saint-Herblain, France; Thomas Powles, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University, Columbus, OH; Osvaldo Arén Frontera, Centro Internacional de Estudios Clinicos, Santiago, Chile; Piotr Tomczak, Szpital Kliniczny Przemienienia Panskiego Uniwersytetu Medycznego, Poznań, Poland; Daniel Castellano, Hospital Universitario 12 de Octubre, Madrid, Spain; Leonard J Appleman, University of Pittsburgh Medical Center, Pittsburgh, PA; Harry Drabkin, Medical University of South Carolina, Charleston, SC; Daniel Vaena, University of Iowa Hospitals and Clinics, Iowa City, IA; Steven Milwee, Jillian Youkstetter, and Julie C. Lougheed, Exelixis, Inc., San Francisco, CA; Sergio Bracarda, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Daniel Vaena
- Bernard Escudier, Institut Gustave Roussy, Villejuif; Stephane Oudard, Hôpital Européen Georges Pompidou, Paris; Frederic Rolland, Centre René Gauducheau Centre de Lutte Contre Le Cancer Nantes, Saint-Herblain, France; Thomas Powles, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University, Columbus, OH; Osvaldo Arén Frontera, Centro Internacional de Estudios Clinicos, Santiago, Chile; Piotr Tomczak, Szpital Kliniczny Przemienienia Panskiego Uniwersytetu Medycznego, Poznań, Poland; Daniel Castellano, Hospital Universitario 12 de Octubre, Madrid, Spain; Leonard J Appleman, University of Pittsburgh Medical Center, Pittsburgh, PA; Harry Drabkin, Medical University of South Carolina, Charleston, SC; Daniel Vaena, University of Iowa Hospitals and Clinics, Iowa City, IA; Steven Milwee, Jillian Youkstetter, and Julie C. Lougheed, Exelixis, Inc., San Francisco, CA; Sergio Bracarda, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Steven Milwee
- Bernard Escudier, Institut Gustave Roussy, Villejuif; Stephane Oudard, Hôpital Européen Georges Pompidou, Paris; Frederic Rolland, Centre René Gauducheau Centre de Lutte Contre Le Cancer Nantes, Saint-Herblain, France; Thomas Powles, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University, Columbus, OH; Osvaldo Arén Frontera, Centro Internacional de Estudios Clinicos, Santiago, Chile; Piotr Tomczak, Szpital Kliniczny Przemienienia Panskiego Uniwersytetu Medycznego, Poznań, Poland; Daniel Castellano, Hospital Universitario 12 de Octubre, Madrid, Spain; Leonard J Appleman, University of Pittsburgh Medical Center, Pittsburgh, PA; Harry Drabkin, Medical University of South Carolina, Charleston, SC; Daniel Vaena, University of Iowa Hospitals and Clinics, Iowa City, IA; Steven Milwee, Jillian Youkstetter, and Julie C. Lougheed, Exelixis, Inc., San Francisco, CA; Sergio Bracarda, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Jillian Youkstetter
- Bernard Escudier, Institut Gustave Roussy, Villejuif; Stephane Oudard, Hôpital Européen Georges Pompidou, Paris; Frederic Rolland, Centre René Gauducheau Centre de Lutte Contre Le Cancer Nantes, Saint-Herblain, France; Thomas Powles, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University, Columbus, OH; Osvaldo Arén Frontera, Centro Internacional de Estudios Clinicos, Santiago, Chile; Piotr Tomczak, Szpital Kliniczny Przemienienia Panskiego Uniwersytetu Medycznego, Poznań, Poland; Daniel Castellano, Hospital Universitario 12 de Octubre, Madrid, Spain; Leonard J Appleman, University of Pittsburgh Medical Center, Pittsburgh, PA; Harry Drabkin, Medical University of South Carolina, Charleston, SC; Daniel Vaena, University of Iowa Hospitals and Clinics, Iowa City, IA; Steven Milwee, Jillian Youkstetter, and Julie C. Lougheed, Exelixis, Inc., San Francisco, CA; Sergio Bracarda, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Julie C Lougheed
- Bernard Escudier, Institut Gustave Roussy, Villejuif; Stephane Oudard, Hôpital Européen Georges Pompidou, Paris; Frederic Rolland, Centre René Gauducheau Centre de Lutte Contre Le Cancer Nantes, Saint-Herblain, France; Thomas Powles, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University, Columbus, OH; Osvaldo Arén Frontera, Centro Internacional de Estudios Clinicos, Santiago, Chile; Piotr Tomczak, Szpital Kliniczny Przemienienia Panskiego Uniwersytetu Medycznego, Poznań, Poland; Daniel Castellano, Hospital Universitario 12 de Octubre, Madrid, Spain; Leonard J Appleman, University of Pittsburgh Medical Center, Pittsburgh, PA; Harry Drabkin, Medical University of South Carolina, Charleston, SC; Daniel Vaena, University of Iowa Hospitals and Clinics, Iowa City, IA; Steven Milwee, Jillian Youkstetter, and Julie C. Lougheed, Exelixis, Inc., San Francisco, CA; Sergio Bracarda, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Sergio Bracarda
- Bernard Escudier, Institut Gustave Roussy, Villejuif; Stephane Oudard, Hôpital Européen Georges Pompidou, Paris; Frederic Rolland, Centre René Gauducheau Centre de Lutte Contre Le Cancer Nantes, Saint-Herblain, France; Thomas Powles, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University, Columbus, OH; Osvaldo Arén Frontera, Centro Internacional de Estudios Clinicos, Santiago, Chile; Piotr Tomczak, Szpital Kliniczny Przemienienia Panskiego Uniwersytetu Medycznego, Poznań, Poland; Daniel Castellano, Hospital Universitario 12 de Octubre, Madrid, Spain; Leonard J Appleman, University of Pittsburgh Medical Center, Pittsburgh, PA; Harry Drabkin, Medical University of South Carolina, Charleston, SC; Daniel Vaena, University of Iowa Hospitals and Clinics, Iowa City, IA; Steven Milwee, Jillian Youkstetter, and Julie C. Lougheed, Exelixis, Inc., San Francisco, CA; Sergio Bracarda, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| | - Toni K Choueiri
- Bernard Escudier, Institut Gustave Roussy, Villejuif; Stephane Oudard, Hôpital Européen Georges Pompidou, Paris; Frederic Rolland, Centre René Gauducheau Centre de Lutte Contre Le Cancer Nantes, Saint-Herblain, France; Thomas Powles, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; Robert J. Motzer, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University, Columbus, OH; Osvaldo Arén Frontera, Centro Internacional de Estudios Clinicos, Santiago, Chile; Piotr Tomczak, Szpital Kliniczny Przemienienia Panskiego Uniwersytetu Medycznego, Poznań, Poland; Daniel Castellano, Hospital Universitario 12 de Octubre, Madrid, Spain; Leonard J Appleman, University of Pittsburgh Medical Center, Pittsburgh, PA; Harry Drabkin, Medical University of South Carolina, Charleston, SC; Daniel Vaena, University of Iowa Hospitals and Clinics, Iowa City, IA; Steven Milwee, Jillian Youkstetter, and Julie C. Lougheed, Exelixis, Inc., San Francisco, CA; Sergio Bracarda, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy; and Toni K. Choueiri, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
26
|
Lam HM, Nguyen HM, Corey E. Generation of Prostate Cancer Patient-Derived Xenografts to Investigate Mechanisms of Novel Treatments and Treatment Resistance. Methods Mol Biol 2018; 1786:1-27. [PMID: 29786784 DOI: 10.1007/978-1-4939-7845-8_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Treatment advances lead to survival benefits of patients with advanced prostate cancer. These treatments are highly efficacious in a subset of patients; however, similarly to other cancers, after initial responses the tumors develop resistance (acquired resistance) and the patients succumb to the disease. Furthermore, there is a subset of patients who do not respond to the treatment at all (de novo resistance). Preclinical testing using patient-derived xenografts (PDXs) has led to successful drug development, and PDXs will continue to provide valuable resources to generate clinically relevant data with translational potential. PDXs demonstrate tumor heterogeneity observed in patients, preserve tumor-microenvironment architecture, and provide clinically relevant treatment responses. In view of the evolving biology of the advanced prostate cancer associated with new treatments, PDXs representing these new tumor phenotypes are urgently needed for the study of treatment responses and resistance. In this chapter, we describe methodologies used to establish prostate cancer PDXs and use of these PDXs to study de novo and acquired resistance.
Collapse
Affiliation(s)
- Hung-Ming Lam
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Holly M Nguyen
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
27
|
Pan T, Lin SC, Yu KJ, Yu G, Song JH, Lewis VO, Bird JE, Moon B, Lin PP, Tannir NM, Jonasch E, Wood CG, Gallick GE, Yu-Lee LY, Lin SH, Satcher RL. BIGH3 Promotes Osteolytic Lesions in Renal Cell Carcinoma Bone Metastasis by Inhibiting Osteoblast Differentiation. Neoplasia 2017; 20:32-43. [PMID: 29190493 PMCID: PMC5711998 DOI: 10.1016/j.neo.2017.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/01/2017] [Accepted: 11/02/2017] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND: Bone metastasis is common in renal cell carcinoma (RCC), and the lesions are mainly osteolytic. The mechanism of bone destruction in RCC bone metastasis is unknown. METHODS: We used a direct intrafemur injection of mice with bone-derived 786-O RCC cells (Bo-786) as an in vivo model to study if inhibition of osteoblast differentiation is involved in osteolytic bone lesions in RCC bone metastasis. RESULTS: We showed that bone-derived Bo-786 cells induced osteolytic bone lesions in the femur of mice. We examined the effect of conditioned medium of Bo-786 cells (Bo-786 CM) on both primary mouse osteoblasts and MC3T3-E1 preosteoblasts and found that Bo-786 CM inhibited osteoblast differentiation. Secretome analysis of Bo-786 CM revealed that BIGH3 (Beta ig h3 protein), also known as TGFBI (transforming growth factor beta-induced protein), is highly expressed. We generated recombinant BIGH3 and found that BIGH3 inhibited osteoblast differentiation in vitro. In addition, CM from Bo-786 BIGH3 knockdown cells (786-BIGH3 KD) reduced the inhibition of osteoblast differentiation compared to CM from vector control. Intrafemural injection of mice with 786-BIGH3 KD cells showed a reduction in osteolytic bone lesions compared to vector control. Immunohistochemical staining of 18 bone metastasis specimens from human RCC showed strong BIGH3 expression in 11/18 (61%) and moderate BIGH3 expression in 7/18 (39%) of the specimens. CONCLUSIONS: These results suggest that suppression of osteoblast differentiation by BIGH3 is one of the mechanisms that enhance osteolytic lesions in RCC bone metastasis, and raise the possibilty that treatments that increase bone formation may improve therapy outcomes.
Collapse
Affiliation(s)
- Tianhong Pan
- Department of Orthopedic Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Song-Chang Lin
- Department of Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Kai-Jie Yu
- Department of Urology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA; Division of Urology, Department of Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan; Department of Chemical Engineering and Biotechnology and Graduate Institute of Biochemical and Biomedical Engineering, National Taipei University of Technology, Taipei, Taiwan
| | - Guoyu Yu
- Department of Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Jian H Song
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Valerae O Lewis
- Department of Orthopedic Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Justin E Bird
- Department of Orthopedic Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Bryan Moon
- Department of Orthopedic Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Patrick P Lin
- Department of Orthopedic Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Nizar M Tannir
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Eric Jonasch
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher G Wood
- Department of Urology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Gary E Gallick
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Li-Yuan Yu-Lee
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Sue-Hwa Lin
- Department of Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA; Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| | - Robert L Satcher
- Department of Orthopedic Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
28
|
Schöffski P, Gordon M, Smith DC, Kurzrock R, Daud A, Vogelzang NJ, Lee Y, Scheffold C, Shapiro GI. Phase II randomised discontinuation trial of cabozantinib in patients with advanced solid tumours. Eur J Cancer 2017; 86:296-304. [PMID: 29059635 DOI: 10.1016/j.ejca.2017.09.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 09/14/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Cabozantinib is an inhibitor of tyrosine kinases, including MET, vascular endothelial growth factor receptor, AXL and RET. This multi-cohort phase II randomised discontinuation trial explored anticancer activity of cabozantinib in nine tumour types. PATIENTS AND METHODS Cabozantinib was administered (100 mg, once daily) to patients with advanced, recurrent or metastatic cancers. Those with stable disease at week 12 were randomised 1:1 to cabozantinib or placebo. Primary end-points were objective response rate (ORR) at week 12 and progression-free survival (PFS) in the randomised phase. RESULTS A total of 526 patients were enrolled. The highest ORR was observed in ovarian cancer (OC) (21.7%); the largest PFS benefit was observed in castration-resistant prostate cancer (CRPC) (median 5.5 versus 1.4 months for placebo; hazard ratio 0.14, 95% confidence interval: 0.04, 0.52). Disease control rates were >40% for CRPC, OC, melanoma, metastatic breast cancer (MBC), hepatocellular carcinoma (HCC) and non-small cell lung cancer. Median duration of response ranged from 3.3 (MBC) to 11.2 months (OC). Encouraging efficacy results and symptomatic improvements prompted early suspension of the randomised stage and conversion to open-label non-randomised expansion cohorts. Dose reductions to manage adverse events (AEs) occurred in 48.7% of patients. The most frequent grade III-IV AEs were fatigue (12.4%), diarrhoea (10.5%), hypertension (10.5%) and palmar-plantar erythrodysesthesia syndrome (8.7%). CONCLUSIONS Clinical antitumour activity of cabozantinib was observed in a subset of tumour types: CRPC and OC were evaluated further in expansion cohorts. Phase III programs were initiated in CRPC and HCC. Interpretation of efficacy outcomes was limited by early termination of the randomised portion of the trial. TRIAL REGISTRATION NUMBER NCT00940225.
Collapse
Affiliation(s)
- Patrick Schöffski
- Department of General Medical Oncology, Leuven Cancer Institute, University Hospital Leuven, Leuven, Belgium.
| | | | - David C Smith
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI, USA.
| | - Razelle Kurzrock
- Moores Cancer Center, University of California, San Diego, San Diego, CA, USA.
| | - Adil Daud
- Medical Center at Parnassus, University of California, San Francisco, San Francisco, CA, USA.
| | - Nicholas J Vogelzang
- Comprehensive Cancer Centers of Nevada, US Oncology Research, Las Vegas, NV, USA.
| | - Yihua Lee
- Exelixis, Inc., South San Francisco, CA, USA.
| | | | - Geoffrey I Shapiro
- Early Drug Development Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
29
|
Yu KJ, Li JK, Lee YC, Yu G, Lin SC, Pan T, Satcher RL, Titus MA, Yu-Lee LY, Weng WH, Gallick GE, Lin SH. Cabozantinib-induced osteoblast secretome promotes survival and migration of metastatic prostate cancer cells in bone. Oncotarget 2017; 8:74987-75006. [PMID: 29088840 PMCID: PMC5650395 DOI: 10.18632/oncotarget.20489] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 07/14/2017] [Indexed: 12/25/2022] Open
Abstract
Therapies that target cancer cells may have unexpected effects on the tumor microenvironment that affects therapy outcomes or render therapy resistance. Prostate cancer (PCa) bone metastasis is uniquely associated with osteoblastic bone lesions and treatment with cabozantinib, a VEGFR-2 and MET inhibitor, leads to a reduction in number and/or intensity of lesions on bone scans. However, resistance to cabozantinib therapy inevitably occurs. We examined the effect of cabozantinib on osteoblast differentiation and secretion in the context of therapy resistance. We showed that primary mouse osteoblasts express VEGFR2 and MET and cabozantinib treatment decreased osteoblast proliferation but enhanced their differentiation. A genome-wide analysis of transcriptional responses of osteoblasts to cabozantinib identified a set of genes accounting for inhibition of proliferation and stimulation of differentiation, and a spectrum of secreted proteins induced by cabozantinib, including pappalysin, IGFBP2, WNT 16, and DKK1. We determined that these proteins were upregulated in the conditioned medium of cabozantinib-treated osteoblasts (CBZ-CM) compared to control CM. Treatment of C4-2B4 or PC3-mm2 PCa cells with CBZ-CM increased the anchorage-independent growth and migration of these PCa cells compared to cells treated with control CM. These results suggest that the effect of cabozantinib on the tumor microenvironment may increase tumor cell survival and cause therapy resistance.
Collapse
Affiliation(s)
- Kai-Jie Yu
- Department of Translational Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA.,Division of Urology, Department of Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan.,Department of Chemical Engineering and Biotechnology and Graduate Institute of Biochemical and Biomedical Engineering, National Taipei University of Technology, Taipei, Taiwan
| | - Jeffrey K Li
- Department of Translational Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Yu-Chen Lee
- Department of Translational Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Guoyu Yu
- Department of Translational Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Song-Chang Lin
- Department of Translational Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Tianhong Pan
- Department of Orthopedic Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Robert L Satcher
- Department of Orthopedic Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mark A Titus
- Department of Genitourinary Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Li-Yuan Yu-Lee
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Wen Hui Weng
- Department of Chemical Engineering and Biotechnology and Graduate Institute of Biochemical and Biomedical Engineering, National Taipei University of Technology, Taipei, Taiwan
| | - Gary E Gallick
- Department of Genitourinary Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA.,The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas, USA
| | - Sue-Hwa Lin
- Department of Translational Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA.,Department of Genitourinary Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA.,The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas, USA
| |
Collapse
|
30
|
Abdelaziz A, Vaishampayan U. Cabozantinib for Renal Cell Carcinoma: Current and Future Paradigms. Curr Treat Options Oncol 2017; 18:18. [PMID: 28286925 DOI: 10.1007/s11864-017-0444-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
OPINION STATEMENT Cabozantinib was approved by the FDA in April 2016 for the treatment of advanced renal cancer, pretreated with at least one prior antiangiogenic therapy. This is the first agent in the therapy of kidney cancer to show a statistically significant improvement in all three endpoints of clinical efficacy, response rate, progression free survival, and overall survival (OS), in a phase III randomized trial. The reporting of METEOR coincided with that of the Checkmate 025 study which randomized similarly eligible patients to receive nivolumab or everolimus 10 mg daily. As the drug development has occurred in parallel for cabozantinib and nivolumab, no evidence exists for decision making regarding optimal sequencing of these agents. A third option of lenvatinib and everolimus was also rapidly approved based on a phase II randomized trial demonstrating promising magnitude of improvement in response, progression-free survival (PFS), and OS. The differences in toxicity profiles, duration and toxicities of prior therapy, presence of brain metastases, concomitant immunosuppressive therapies, or autoimmune conditions are the factors that are taken into account when choosing therapy. The patients who have demonstrated response, prolonged clinical benefit and tolerability, and with anti-VEGF therapy are likely to benefit from continued antiangiogenic activity combined with MET and HGF inhibition with cabozantinib at progression. The patients who have intolerance or poor response to anti-VEGF TKI should be switched to nivolumab as the preferential therapy of choice. Clearly, better predictors are required to aid in guiding therapeutic decisions. The CABOSUN trial will likely shift the entire paradigm. The CABOSUN trial demonstrated superior PFS and response rates favoring cabozantinib as compared to sunitinib in untreated, intermediate, or poor-risk RCC and can be predicted to become the front-line therapy of choice. Immune-based regimens such as the combinations of nivolumab + ipilimumab and bevacizumab + atezolizumab have completed phase III trials, comparing to sunitinib, and results are awaited. In the future, a similar clinical dilemma will be shifted to the front-line therapy and the nuances of trial eligibility, and patient comorbidities will remain important factors. Optimal sequencing and predictive biomarkers are the questions that need to be incorporated in future clinical trials within RCC.
Collapse
Affiliation(s)
- Ahmed Abdelaziz
- Department of Oncology, Wayne State University/Barbara Ann Karmanos Cancer Center, 4100 John R St, Detroit, MI, 48201, USA
| | - Ulka Vaishampayan
- Department of Oncology, Wayne State University/Barbara Ann Karmanos Cancer Center, 4100 John R St, Detroit, MI, 48201, USA.
| |
Collapse
|
31
|
Abstract
INTRODUCTION Cabozantinib is a small molecule tyrosine kinase inhibitor that initially showed activity in medullary thyroid cancer and was recently approved by the Food and Drug Administration for the treatment of metastatic renal cell carcinoma after progression on first line therapy. Areas covered: In the METEOR trial, cabozantinib demonstrated significantly improved efficacy in all three endpoints; response rates, progression free survival and overall survival in a randomized trial with everolimus as an active comparator. Cabozantinib also showed activity in the front line setting in RCC within the CABOSUN trial. The study randomized untreated metastatic RCC patients to either cabozantinib or sunitinib and the former showed improved progression free survival which was the primary endpoint. The future holds promise for indications in other malignancies, given the preliminary efficacy and unique mechanism of action of cabozantinib. In this review we address the mechanism of action, pharmacodynamics and pharmacokinetics of cabozantinib, and also review the development pathway of this agent in the treatment of advanced renal cell carcinoma. The potential benefit in specific patient populations, such as poor risk patients and bone metastases subgroups is also discussed. Expert commentary: The clinical applications of cabozantinib will be addressed within the context of the current competitive therapeutic landscape of RCC.
Collapse
Affiliation(s)
- Ahmed Abdelaziz
- a Department of Oncology , Wayne State University/Barbara Ann Karmanos Cancer Institute , Detroit , MI , USA
| | - Ulka Vaishampayan
- a Department of Oncology , Wayne State University/Barbara Ann Karmanos Cancer Institute , Detroit , MI , USA
| |
Collapse
|
32
|
Cochin V, Gross-Goupil M, Ravaud A, Godbert Y, Le Moulec S. [Cabozantinib: Mechanism of action, efficacy and indications]. Bull Cancer 2017; 104:393-401. [PMID: 28477875 DOI: 10.1016/j.bulcan.2017.03.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 03/15/2017] [Accepted: 03/24/2017] [Indexed: 11/28/2022]
Abstract
Cabozantinib is an oral multiple tyrosine kinase receptor inhibitor (ITK): VEGFR2, c-MET and RET. Inhibition of VEGFR and c-MET decrease resistance of VEGFR inhibitor via c-MET axis. Cabozantinib improve progression-free survival (PFS) in progressive metastatic medullary thyroid cancer (MTC): 4 months in the placebo group and 11.2 months in the cabozantinib group (P<0.001) in all patient subgroups including those with or without prior ITK and RET mutation status. Cabozantinib increased overall survival (OS) compared with everolimus in patients with advanced renal cell carcinoma who progressed after previous VEGFR ITK treatment: 21.4 months in cabozantinib group and 16.5 months in everolimus group (P<0.0003). Cabozantinib obtained the AMM for the treatment of progressive metastatic MTC and advanced renal cell carcinoma. Cabozantinib is a new option in the treatment of MTC by inclusion in therapeutic trials (no payment in this indication) and advanced renal cell carcinoma (hospital delivery). Its tolerance is similar to anti-angiogenic therapies and justifies an optimal management of the secondary effect.
Collapse
Affiliation(s)
- Valérie Cochin
- Centre hospitalo-universitaire de Bordeaux, hôpital Saint-André, 01, rue Jean-Burguet, 33075 Bordeaux cedex, France.
| | - Marine Gross-Goupil
- Centre hospitalo-universitaire de Bordeaux, hôpital Saint-André, 01, rue Jean-Burguet, 33075 Bordeaux cedex, France
| | - Alain Ravaud
- Centre hospitalo-universitaire de Bordeaux, hôpital Saint-André, 01, rue Jean-Burguet, 33075 Bordeaux cedex, France; Université de Bordeaux, 146, rue Léo-Saignat, 33076 Bordeaux cedex, France
| | - Yann Godbert
- Institut Bergonié, centre de lutte contre le cancer de Bordeaux, 229, cours de l'Argonne, 33076 Bordeaux cedex, France
| | - Sylvestre Le Moulec
- Institut Bergonié, centre de lutte contre le cancer de Bordeaux, 229, cours de l'Argonne, 33076 Bordeaux cedex, France
| |
Collapse
|
33
|
Nguyen HM, Vessella RL, Morrissey C, Brown LG, Coleman IM, Higano CS, Mostaghel EA, Zhang X, True LD, Lam H, Roudier M, Lange PH, Nelson PS, Corey E. LuCaP Prostate Cancer Patient-Derived Xenografts Reflect the Molecular Heterogeneity of Advanced Disease an--d Serve as Models for Evaluating Cancer Therapeutics. Prostate 2017; 77:654-671. [PMID: 28156002 PMCID: PMC5354949 DOI: 10.1002/pros.23313] [Citation(s) in RCA: 205] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 01/06/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND Metastatic prostate cancer is a common and lethal disease for which there are no therapies that produce cures or long-term durable remissions. Clinically relevant preclinical models are needed to increase our understanding of biology of this malignancy and to evaluate new agents that might provide effective treatment. Our objective was to establish and characterize patient-derived xenografts (PDXs) from advanced prostate cancer (PC) for investigation of biology and evaluation of new treatment modalities. METHODS Samples of advanced PC obtained from primary prostate cancer obtained at surgery or from metastases collected at time of death were implanted into immunocompromised mice to establish PDXs. Established PDXs were propagated in vivo. Genomic, transcriptomic, and STR profiles were generated. Responses to androgen deprivation and docetaxel in vivo were characterized. RESULTS We established multiple PDXs (LuCaP series), which represent the major genomic and phenotypic features of the disease in humans, including amplification of androgen receptor, PTEN deletion, TP53 deletion and mutation, RB1 loss, TMPRSS2-ERG rearrangements, SPOP mutation, hypermutation due to MSH2/MSH6 genomic aberrations, and BRCA2 loss. The PDX models also exhibit variation in intra-tumoral androgen levels. Our in vivo results show heterogeneity of response to androgen deprivation and docetaxel, standard therapies for advanced PC, similar to the responses of patients to these treatments. CONCLUSIONS The LuCaP PDX series reflects the diverse molecular composition of human castration-resistant PC and allows for hypothesis-driven cause-and-effect studies of mechanisms underlying treatment response and resistance. Prostate 77: 654-671, 2017. © 2017 The Authors. The Prostate Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Holly M. Nguyen
- Department of UrologyUniversity of WashingtonSeattleWashington
| | - Robert L. Vessella
- Department of UrologyUniversity of WashingtonSeattleWashington
- Puget Sound Veteran AdministrationSeattleWashington
| | - Colm Morrissey
- Department of UrologyUniversity of WashingtonSeattleWashington
| | - Lisha G. Brown
- Department of UrologyUniversity of WashingtonSeattleWashington
| | - Ilsa M. Coleman
- Division of Human BiologyFred Hutchinson Cancer Research CenterSeattleWashington
| | - Celestia S. Higano
- Division of Clinical ResearchFred Hutchinson Cancer Research CenterSeattleWashington
- Division of OncologyDepartment of MedicineUniversity of WashingtonSeattleWashington
| | - Elahe A. Mostaghel
- Division of Clinical ResearchFred Hutchinson Cancer Research CenterSeattleWashington
| | - Xiaotun Zhang
- Department of UrologyUniversity of WashingtonSeattleWashington
| | - Lawrence D. True
- Department of PathologyUniversity of WashingtonSeattleWashington
| | - Hung‐Ming Lam
- Department of UrologyUniversity of WashingtonSeattleWashington
| | - Martine Roudier
- Department of UrologyUniversity of WashingtonSeattleWashington
| | - Paul H. Lange
- Department of UrologyUniversity of WashingtonSeattleWashington
| | - Peter S. Nelson
- Department of UrologyUniversity of WashingtonSeattleWashington
- Division of Human BiologyFred Hutchinson Cancer Research CenterSeattleWashington
- Department of PathologyUniversity of WashingtonSeattleWashington
| | - Eva Corey
- Department of UrologyUniversity of WashingtonSeattleWashington
| |
Collapse
|
34
|
Fioramonti M, Santini D, Iuliani M, Ribelli G, Manca P, Papapietro N, Spiezia F, Vincenzi B, Denaro V, Russo A, Tonini G, Pantano F. Cabozantinib targets bone microenvironment modulating human osteoclast and osteoblast functions. Oncotarget 2017; 8:20113-20121. [PMID: 28223547 PMCID: PMC5386748 DOI: 10.18632/oncotarget.15390] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 01/22/2017] [Indexed: 12/20/2022] Open
Abstract
Cabozantinib, a c-MET and vascular endothelial growth factor receptor 2 inhibitor, demonstrated to prolong progression free survival and improve skeletal disease-related endpoints in castration-resistant prostate cancer and in metastatic renal carcinoma. Our purpose is to investigate the direct effect of cabozantinib on bone microenvironment using a total human model of primary osteoclasts and osteoblasts.Osteoclasts were differentiated from monocytes isolated from healthy donors; osteoblasts were derived from human mesenchymal stem cells obtained from bone fragments of orthopedic surgery patients. Osteoclast activity was evaluated by tartrate resistant acid phosphatase (TRAP) staining and bone resorption assays and osteoblast differentiation was detected by alkaline phosphatase and alizarin red staining.Our results show that non-cytotoxic doses of cabozantinib significantly inhibit osteoclast differentiation (p=0.0145) and bone resorption activity (p=0.0252). Moreover, cabozantinib down-modulates the expression of osteoclast marker genes, TRAP (p=0.006), CATHEPSIN K (p=0.004) and Receptor Activator of Nuclear Factor k B (RANK) (p=0.001). Cabozantinib treatment has no effect on osteoblast viability or differentiation, but increases osteoprotegerin mRNA (p=0.015) and protein levels (p=0.004) and down-modulates Receptor Activator of Nuclear Factor k B Ligand (RANKL) at both mRNA (p<0.001) and protein levels (p=0.043). Direct cell-to-cell contact between cabozantinib pre-treated osteoblasts and untreated osteoclasts confirmed the indirect anti-resorptive effect of cabozantinib.We demonstrate that cabozantinib inhibits osteoclast functions "directly" and "indirectly" reducing the RANKL/osteoprotegerin ratio in osteoblasts.
Collapse
Affiliation(s)
- Marco Fioramonti
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Rome, Italy
| | - Daniele Santini
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Rome, Italy
| | - Michele Iuliani
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Rome, Italy
| | - Giulia Ribelli
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Rome, Italy
| | - Paolo Manca
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Rome, Italy
| | - Nicola Papapietro
- Department of Orthopaedics and Trauma Surgery, University Campus Bio-Medico of Rome, Rome, Italy
| | - Filippo Spiezia
- Department of Orthopaedics and Trauma Surgery, University Campus Bio-Medico of Rome, Rome, Italy
| | - Bruno Vincenzi
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Rome, Italy
| | - Vincenzo Denaro
- Department of Orthopaedics and Trauma Surgery, University Campus Bio-Medico of Rome, Rome, Italy
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Giuseppe Tonini
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Rome, Italy
| | - Francesco Pantano
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Rome, Italy
| |
Collapse
|
35
|
Effective combinatorial immunotherapy for castration-resistant prostate cancer. Nature 2017; 543:728-732. [PMID: 28321130 PMCID: PMC5374023 DOI: 10.1038/nature21676] [Citation(s) in RCA: 393] [Impact Index Per Article: 56.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 01/31/2017] [Indexed: 12/12/2022]
Abstract
A significant fraction of advanced prostate cancer (PCa) patients treated with androgen deprivation therapy (ADT) experience relapse with relentless progression to lethal metastatic castration-resistant prostate cancer (mCRPC)1. Immune checkpoint blockade (ICB) using antibodies against cytotoxic-T-lymphocyte-associated protein 4 (CTLA4) or programmed cell death 1/programmed cell death 1 ligand 1 (PD1/PD-L1) generates durable therapeutic responses in a significant subset of patients across a variety of cancer types2. However, mCRPC showed overwhelming de novo resistance to ICB3–5, motivating a search for targeted therapies that overcome this resistance. Myeloid-derived suppressor cells (MDSCs) are known to play important roles in tumor immune evasion6. Circulating MDSC abundance correlates with PSA levels and metastasis in PCa patients7–9. Mouse models of PCa show that MDSCs (CD11b+ Gr1+) promote tumor initiation10 and progression11. These observations prompted us to hypothesize that robust immunotherapy responses in mCRPC may be elicited by the combined actions of ICB agents together with targeted agents that neutralize MDSCs yet preserve T cell function. Here we developed a novel chimeric mouse model of mCRPC to efficiently test combination therapies in an autochthonous setting. Combination of anti-CTLA4 and anti-PD1 engendered only modest efficacy. Targeted therapy against mCRPC-infiltrating MDSCs, using multikinase inhibitors such as cabozantinib and BEZ235, also showed minimal anti-tumor activities. Strikingly, primary and metastatic CRPC showed robust synergistic responses when ICB was combined with MDSC-targeted therapy. Mechanistically, combination therapy efficacy stemmed from the upregulation of IL-1ra and suppression of MDSC-promoting cytokines secreted by PCa cells. These observations illuminate a clinical path hypothesis for combining ICB with MDSC-targeted therapies in the treatment of mCRPC.
Collapse
|
36
|
C-Met/miR-130b axis as novel mechanism and biomarker for castration resistance state acquisition. Oncogene 2017; 36:3718-3728. [PMID: 28192399 DOI: 10.1038/onc.2016.505] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 11/06/2016] [Accepted: 11/29/2016] [Indexed: 12/19/2022]
Abstract
Although a significant subset of prostate tumors remain indolent during the entire life, the advanced forms are still one of the leading cause of cancer-related death. There are not reliable markers distinguishing indolent from aggressive forms. Here we highlighted a new molecular circuitry involving microRNA and coding genes promoting cancer progression and castration resistance. Our preclinical and clinical data demonstrated that c-Met activation increases miR-130b levels, inhibits androgen receptor expression, promotes cancer spreading and resistance to hormone ablation therapy. The relevance of these findings was confirmed on patients' samples and by in silico analysis on an independent patient cohort from Taylor's platform. Data suggest c-Met/miR-130b axis as a new prognostic marker for patients' risk assessment and as an indicator of therapy resistance. Our results propose new biomarkers for therapy decision-making in all phases of the pathology. Data may help identify high-risk patients to be treated with adjuvant therapy together with alternative cure for castration-resistant forms while facilitating the identification of possible patients candidates for anti-Met therapy. In addition, we demonstrated that it is possible to evaluate Met/miR-130b axis expression in exosomes isolated from peripheral blood of surgery candidates and advanced patients offering a new non-invasive tool for active surveillance and therapy monitoring.
Collapse
|
37
|
Shintani T, Kusuhara Y, Daizumoto K, Dondoo TO, Yamamoto H, Mori H, Fukawa T, Nakatsuji H, Fukumori T, Takahashi M, Kanayama H. The Involvement of Hepatocyte Growth Factor-MET-Matrix Metalloproteinase 1 Signaling in Bladder Cancer Invasiveness and Proliferation. Effect of the MET Inhibitor, Cabozantinib (XL184), on Bladder Cancer Cells. Urology 2016; 101:169.e7-169.e13. [PMID: 28013036 DOI: 10.1016/j.urology.2016.12.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 11/09/2016] [Accepted: 12/03/2016] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To clarify the invasive mechanisms of muscle-invasive bladder cancer (BCa) would be useful for the determination of appropriate treatment strategies. We previously showed that hepatocyte growth factor (HGF)-MET signaling is correlated with invasiveness of BCa cells. Here, we investigated the effects of the MET inhibitor, cabozantinib (XL184), on BCa cells. METHODS We first conducted Western blot analysis to investigate MET expression in BCa cell lines. Next, we examined the effect of cabozantinib on their proliferation and invasive abilities using MTT and Matrigel invasion assays, respectively. Invasion assays were performed using the xCELLigence system. Additionally, to investigate the biological function of HGF-MET signaling, we analyzed gene expression profiles and performed real-time polymerase chain reaction analyses of 5637 cells that were cultivated with or without HGF stimulation, with or without cabozantinib. RESULTS MET was highly expressed in 4 of 5 BCa cell lines, and 5637 and T24 cells showed especially high protein expression of MET. Cabozantinib suppressed cell proliferation and invasion (cell index; mock, 1.49 vs HGF, 2.26 vs HGF + XL184, 1.47, P < .05). Gene expression profile analysis indicated that matrix metalloproteinase 1 (MMP1) was significantly elevated at the mRNA level with addition of HGF. Moreover, cabozantinib suppressed HGF-induced MMP1 expression in 5637 T24 cells. CONCLUSIONS These data indicate that cabozantinib suppressed MMP1 expression by blocking HGF-MET signaling and that HGF-MET-MMP1 signaling is involved in the invasiveness and proliferation of BCa cells. These results suggest that cabozantinib might prove useful for future treatment of muscle-invasive BCa.
Collapse
Affiliation(s)
- Terumichi Shintani
- Department of Urology, Institute of Biomedical Sciences, Tokushima University, Graduate School, Tokushima, Japan
| | - Yoshito Kusuhara
- Department of Urology, Institute of Biomedical Sciences, Tokushima University, Graduate School, Tokushima, Japan
| | - Kei Daizumoto
- Department of Urology, Institute of Biomedical Sciences, Tokushima University, Graduate School, Tokushima, Japan
| | - Tsogt-Ochir Dondoo
- Department of Urology, Institute of Biomedical Sciences, Tokushima University, Graduate School, Tokushima, Japan
| | - Hiroki Yamamoto
- Department of Urology, Institute of Biomedical Sciences, Tokushima University, Graduate School, Tokushima, Japan
| | - Hidehisa Mori
- Department of Urology, Institute of Biomedical Sciences, Tokushima University, Graduate School, Tokushima, Japan
| | - Tomoya Fukawa
- Department of Urology, Institute of Biomedical Sciences, Tokushima University, Graduate School, Tokushima, Japan
| | - Hiroyoshi Nakatsuji
- Department of Urology, Institute of Biomedical Sciences, Tokushima University, Graduate School, Tokushima, Japan.
| | - Tomoharu Fukumori
- Department of Urology, Institute of Biomedical Sciences, Tokushima University, Graduate School, Tokushima, Japan
| | - Masayuki Takahashi
- Department of Urology, Institute of Biomedical Sciences, Tokushima University, Graduate School, Tokushima, Japan
| | - Hiroomi Kanayama
- Department of Urology, Institute of Biomedical Sciences, Tokushima University, Graduate School, Tokushima, Japan
| |
Collapse
|
38
|
Lam HM, McMullin R, Nguyen HM, Coleman I, Gormley M, Gulati R, Brown LG, Holt SK, Li W, Ricci DS, Verstraeten K, Thomas S, Mostaghel EA, Nelson PS, Vessella RL, Corey E. Characterization of an Abiraterone Ultraresponsive Phenotype in Castration-Resistant Prostate Cancer Patient-Derived Xenografts. Clin Cancer Res 2016; 23:2301-2312. [PMID: 27993966 DOI: 10.1158/1078-0432.ccr-16-2054] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/09/2016] [Accepted: 12/08/2016] [Indexed: 01/02/2023]
Abstract
Purpose: To identify the molecular signature associated with abiraterone acetate (AA) response and mechanisms underlying AA resistance in castration-resistant prostate cancer patient-derived xenografts (PDXs).Experimental Design: SCID mice bearing LuCaP 136CR, 77CR, 96CR, and 35CR PDXs were treated with AA. Tumor volume and prostate-specific antigen were monitored, and tumors were harvested 7 days after treatment or at end of study for gene expression and immunohistochemical studies.Results: Three phenotypic groups were observed based on AA response. An ultraresponsive phenotype was identified in LuCaP 136CR with significant inhibition of tumor progression and increased survival, intermediate responders LuCaP 77CR and LuCaP 96CR with a modest tumor inhibition and survival benefit, and LuCaP 35CR with minimal tumor inhibition and no survival benefit upon AA treatment. We identified a molecular signature of secreted proteins associated with the AA ultraresponsive phenotype. Upon resistance, AA ultraresponder LuCaP 136CR displayed reduced androgen receptor (AR) signaling and sustainably low nuclear glucocorticoid receptor (nGR) localization, accompanied by steroid metabolism alteration and epithelial-mesenchymal transition phenotype enrichment with increased expression of NF-κB-regulated genes; intermediate and minimal responders maintained sustained AR signaling and increased tumoral nGR localization.Conclusions: We identified a molecular signature of secreted proteins associated with AA ultraresponsiveness and sustained AR/GR signaling upon AA resistance in intermediate or minimal responders. These data will inform development of noninvasive biomarkers predicting AA response and suggest that further inhibition along the AR/GR signaling axis may be effective only in AA-resistant patients who are intermediate or minimal responders. These findings require verification in prospective clinical trials. Clin Cancer Res; 23(9); 2301-12. ©2016 AACR.
Collapse
Affiliation(s)
- Hung-Ming Lam
- Department of Urology, University of Washington, Seattle, Washington.,State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | | | - Holly M Nguyen
- Department of Urology, University of Washington, Seattle, Washington
| | - Ilsa Coleman
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Michael Gormley
- Janssen Research and Development, Spring House, Pennsylvania
| | - Roman Gulati
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Lisha G Brown
- Department of Urology, University of Washington, Seattle, Washington
| | - Sarah K Holt
- Department of Urology, University of Washington, Seattle, Washington
| | - Weimin Li
- Janssen Research and Development, Spring House, Pennsylvania
| | | | | | - Shibu Thomas
- Janssen Research and Development, Spring House, Pennsylvania
| | - Elahe A Mostaghel
- Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Medicine, University of Washington, Seattle, Washington
| | - Peter S Nelson
- Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Medicine, University of Washington, Seattle, Washington
| | - Robert L Vessella
- Department of Urology, University of Washington, Seattle, Washington.,Department of Veterans Affairs Medical Center, Seattle, Washington
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington.
| |
Collapse
|
39
|
Abstract
Cabozantinib inhibits a variety of cellular receptors including VEGFR1-3, MET, AXL, RET, FLT3 and KIT. These signaling pathways have been shown to be important in genitourinary malignancies. Along its clinical development, it has shown most activity in advanced renal cell carcinoma; the METEOR study compared cabozantinib to everolimus and showed clinically and statistically significant improvements in both progression-free survival and overall survival. Herein, we review the development of cabozantinib in the genitourinary malignancies of renal cell carcinoma, prostate adenocarcinoma and urothelial carcinoma.
Collapse
Affiliation(s)
- Tian Zhang
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Se Eun Park
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Cierra Hong
- Duke University School of Medicine, Durham, NC 27710, USA
| | - Daniel J George
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA.,Division of Urology, Departments of Medicine & Surgery, Duke Cancer Institute, Duke University Medical Center, DUMC 103861, Durham, NC 27710, USA
| |
Collapse
|
40
|
Hirata T, Park SC, Muldong MT, Wu CN, Yamaguchi T, Strasner A, Raheem O, Kumon H, Sah RL, Cacalano NA, Jamieson CHM, Kane CJ, Masuda K, Kulidjian AA, Jamieson CAM. Specific bone region localization of osteolytic versus osteoblastic lesions in a patient-derived xenograft model of bone metastatic prostate cancer. Asian J Urol 2016; 3:229-239. [PMID: 29264191 PMCID: PMC5730873 DOI: 10.1016/j.ajur.2016.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 08/26/2016] [Accepted: 08/26/2016] [Indexed: 01/30/2023] Open
Abstract
Objective Bone metastasis occurs in up to 90% of men with advanced prostate cancer and leads to fractures, severe pain and therapy-resistance. Bone metastases induce a spectrum of types of bone lesions which can respond differently to therapy even within individual prostate cancer patients. Thus, the special environment of the bone makes the disease more complicated and incurable. A model in which bone lesions are reproducibly induced that mirrors the complexity seen in patients would be invaluable for pre-clinical testing of novel treatments. The microstructural changes in the femurs of mice implanted with PCSD1, a new patient-derived xenograft from a surgical prostate cancer bone metastasis specimen, were determined. Methods Quantitative micro-computed tomography (micro-CT) and histological analyses were performed to evaluate the effects of direct injection of PCSD1 cells or media alone (Control) into the right femurs of Rag2−/−γc−/− male mice. Results Bone lesions formed only in femurs of mice injected with PCSD1 cells. Bone volume (BV) was significantly decreased at the proximal and distal ends of the femurs (p < 0.01) whereas BV (p < 0.05) and bone shaft diameter (p < 0.01) were significantly increased along the femur shaft. Conclusion PCSD1 cells reproducibly induced bone loss leading to osteolytic lesions at the ends of the femur, and, in contrast, induced aberrant bone formation leading to osteoblastic lesions along the femur shaft. Therefore, the interaction of PCSD1 cells with different bone region-specific microenvironments specified the type of bone lesion. Our approach can be used to determine if different bone regions support more therapy resistant tumor growth, thus, requiring novel treatments.
Collapse
Affiliation(s)
- Takeshi Hirata
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Seung Chol Park
- Department of Urology, Wonkwang University School of Medicine and Hospital, Iksan, South Korea
| | - Michelle T Muldong
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.,Department of Urology, University of California, San Diego, La Jolla, CA, USA.,Department of Surgery, University of California, San Diego, La Jolla, CA, USA
| | - Christina N Wu
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Tomonori Yamaguchi
- Department of Orthopaedic Surgery, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Amy Strasner
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.,Department of Urology, University of California, San Diego, La Jolla, CA, USA.,Department of Surgery, University of California, San Diego, La Jolla, CA, USA
| | - Omer Raheem
- Department of Urology, University of California, San Diego, La Jolla, CA, USA.,Department of Surgery, University of California, San Diego, La Jolla, CA, USA
| | - Hiromi Kumon
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Robert L Sah
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Nicholas A Cacalano
- Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Catriona H M Jamieson
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Christopher J Kane
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.,Department of Urology, University of California, San Diego, La Jolla, CA, USA.,Department of Surgery, University of California, San Diego, La Jolla, CA, USA
| | - Koichi Masuda
- Department of Orthopaedic Surgery, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Anna A Kulidjian
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.,Department of Orthopaedic Surgery, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Christina A M Jamieson
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.,Department of Urology, University of California, San Diego, La Jolla, CA, USA.,Department of Surgery, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
41
|
Smith M, De Bono J, Sternberg C, Le Moulec S, Oudard S, De Giorgi U, Krainer M, Bergman A, Hoelzer W, De Wit R, Bögemann M, Saad F, Cruciani G, Thiery-Vuillemin A, Feyerabend S, Miller K, Houédé N, Hussain S, Lam E, Polikoff J, Stenzl A, Mainwaring P, Ramies D, Hessel C, Weitzman A, Fizazi K. Phase III Study of Cabozantinib in Previously Treated Metastatic Castration-Resistant Prostate Cancer: COMET-1. J Clin Oncol 2016; 34:3005-13. [DOI: 10.1200/jco.2015.65.5597] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose Cabozantinib is an inhibitor of kinases, including MET and vascular endothelial growth factor receptors, and has shown activity in men with previously treated metastatic castration-resistant prostate cancer (mCRPC). This blinded phase III trial compared cabozantinib with prednisone in patients with mCRPC. Patients and Methods Men with progressive mCRPC after docetaxel and abiraterone and/or enzalutamide were randomly assigned at a two-to-one ratio to cabozantinib 60 mg once per day or prednisone 5 mg twice per day. The primary end point was overall survival (OS). Bone scan response (BSR) at week 12 as assessed by independent review committee was the secondary end point; radiographic progression-free survival (rPFS) and effects on circulating tumor cells (CTCs), bone biomarkers, serum prostate-specific antigen (PSA), and symptomatic skeletal events (SSEs) were exploratory assessments. Results A total of 1,028 patients were randomly assigned to cabozantinib (n = 682) or prednisone (n = 346). Median OS was 11.0 months with cabozantinib and 9.8 months with prednisone (hazard ratio, 0.90; 95% CI, 0.76 to 1.06; stratified log-rank P = .213). BSR at week 12 favored cabozantinib (42% v 3%; stratified Cochran-Mantel-Haenszel P < .001). rPFS was improved in the cabozantinib group (median, 5.6 v 2.8 months; hazard ratio, 0.48; 95% CI, 0.40 to 0.57; stratified log-rank P < .001). Cabozantinib was associated with improvements in CTC conversion, bone biomarkers, and post–random assignment incidence of SSEs but not PSA outcomes. Grade 3 to 4 adverse events and discontinuations because of adverse events were higher with cabozantinib than with prednisone (71% v 56% and 33% v 12%, respectively). Conclusion Cabozantinib did not significantly improve OS compared with prednisone in heavily treated patients with mCRPC and progressive disease after docetaxel and abiraterone and/or enzalutamide. Cabozantinib had some activity in improving BSR, rPFS, SSEs, CTC conversions, and bone biomarkers but not PSA outcomes.
Collapse
Affiliation(s)
- Matthew Smith
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Johann De Bono
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Cora Sternberg
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Sylvestre Le Moulec
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Stéphane Oudard
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Ugo De Giorgi
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Michael Krainer
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Andries Bergman
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Wolfgang Hoelzer
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Ronald De Wit
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Martin Bögemann
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Fred Saad
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Giorgio Cruciani
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Antoine Thiery-Vuillemin
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Susan Feyerabend
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Kurt Miller
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Nadine Houédé
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Syed Hussain
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Elaine Lam
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Jonathan Polikoff
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Arnulf Stenzl
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Paul Mainwaring
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - David Ramies
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Colin Hessel
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Aaron Weitzman
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| | - Karim Fizazi
- Matthew Smith, Massachusetts General Hospital, Boston, MA; Johann De Bono, Royal Marsden Hospital, Sutton; Syed Hussain, University of Liverpool, Liverpool, United Kingdom; Cora Sternberg, San Camillo and Forlanini Hospitals, Rome; Ugo De Giorgi, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori–Istituto di Ricovero e Cura a Carattere Scientifico, Meldola; Giorgio Cruciani, Istituto Tumori Romagna, Lugo di Romagna, Italy; Sylvestre Le Moulec, Hôpital d’Instruction des Armées Val-de-Grâce
| |
Collapse
|
42
|
Modena A, Massari F, Ciccarese C, Brunelli M, Santoni M, Montironi R, Martignoni G, Tortora G. Targeting Met and VEGFR Axis in Metastatic Castration-Resistant Prostate Cancer: ‘Game Over’? Target Oncol 2016; 11:431-46. [DOI: 10.1007/s11523-015-0412-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
43
|
Haider MT, Hunter KD, Robinson SP, Graham TJ, Corey E, Dear TN, Hughes R, Brown NJ, Holen I. Rapid modification of the bone microenvironment following short-term treatment with Cabozantinib in vivo. Bone 2015; 81:581-592. [PMID: 26279137 PMCID: PMC4768060 DOI: 10.1016/j.bone.2015.08.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 07/18/2015] [Accepted: 08/04/2015] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Bone metastasis remains incurable with treatment restricted to palliative care. Cabozantinib (CBZ) is targeted against multiple receptor tyrosine kinases involved in tumour pathobiology, including hepatocyte growth factor receptor (MET) and vascular endothelial growth factor receptor 2 (VEGFR-2). CBZ has demonstrated clinical activity in advanced prostate cancer with resolution of lesions visible on bone scans, implicating a potential role of the bone microenvironment as a mediator of CBZ effects. We characterised the effects of short-term administration of CBZ on bone in a range of in vivo models to determine how CBZ affects bone in the absence of tumour. METHODS Studies were performed in a variety of in vivo models including male and female BALB/c nude mice (age 6-17-weeks). Animals received CBZ (30 mg/kg, 5× weekly) or sterile H2O control for 5 or 10 days. Effects on bone integrity (μCT), bone cell activity (PINP, TRAP ELISA), osteoblast and osteoclast number/mm trabecular bone surface, area of epiphyseal growth plate cartilage, megakaryocyte numbers and bone marrow composition were assessed. Effects of longer-term treatment (15-day & 6-week administration) were assessed in male NOD/SCID and beige SCID mice. RESULTS CBZ treatment had significant effects on the bone microenvironment, including reduced osteoclast and increased osteoblast numbers compared to control. Trabecular bone structure was altered after 8 administrations. A significant elongation of the epiphyseal growth plate, in particular the hypertrophic chondrocyte zone, was observed in all CBZ treated animals irrespective of administration schedule. Both male and female BALB/c nude mice had increased megakaryocyte numbers/mm(2) tissue after 10-day CBZ treatment, in addition to vascular ectasia, reduced bone marrow cellularity and extravasation of red blood cells into the extra-vascular bone marrow. All CBZ-induced effects were transient and rapidly lost following cessation of treatment. CONCLUSION Short-term administration of CBZ induces rapid, reversible effects on the bone microenvironment in vivo highlighting a potential role in mediating treatment responses.
Collapse
Affiliation(s)
| | - Keith D Hunter
- School of Clinical Dentistry, University of Sheffield, Sheffield, UK.
| | - Simon P Robinson
- CR-UK Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, Sutton, Surrey, UK.
| | - Timothy J Graham
- CR-UK Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, Sutton, Surrey, UK.
| | - Eva Corey
- Department of Urology, University of Washington Medical Center, Seattle, WA, USA.
| | - T Neil Dear
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
| | - Russell Hughes
- Department of Oncology, University of Sheffield, Sheffield, UK.
| | - Nicola J Brown
- Department of Oncology, University of Sheffield, Sheffield, UK.
| | - Ingunn Holen
- Department of Oncology, University of Sheffield, Sheffield, UK.
| |
Collapse
|
44
|
Doran MG, Spratt DE, Wongvipat J, Ulmert D, Carver BS, Sawyers CL, Evans MJ. Cabozantinib resolves bone scans in tumor-naïve mice harboring skeletal injuries. Mol Imaging 2015; 13. [PMID: 25248353 DOI: 10.2310/7290.2014.00026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The receptor tyrosine kinase inhibitor cabozantinib (XL184, BMS-907351 Cometriq) has displayed impressive clinical activity against several indications, culminating in its recent approval for medullary thyroid cancer. Among malignancies with tropism for the bone (prostate, breast), one striking feature of early clinical reports about this drug has been the rapid and complete resolution of bone scans, a phenomenon almost never observed even among therapies already shown to confer survival benefit. In castration-resistant prostate cancer, not all conventional response indicators change as dramatically posttreatment, raising the possibility that cabozantinib may impair the ability of bone-seeking radionuclides to integrate within the remodeling bone. To test this hypothesis, we surgically induced bone remodeling via physical insult in non-tumor-bearing mice and performed 18F-sodium fluoride (18F-NaF) positron emission tomographic (PET) and technetium 99m-methylene diphosphonate (99mTc-MDP) single-photon emission computed tomographic (SPECT) scans pre- and posttreatment with cabozantinib and related inhibitors. A consistent reduction in the accumulation of either radiotracer at the site of bone remodeling was observed in animals treated with cabozantinib. Given that cabozantinib is known to inhibit several receptor tyrosine kinases, we drugged animals with various permutations of more selective inhibitors to attempt to refine the molecular basis of bone scan resolution. Neither the vascular endothelial growth factor receptor (VEGFR) inhibitor axitinib, the MET inhibitor crizotinib, nor the combination was capable of inhibiting 18F-NaF accumulation at known bioactive doses. In summary, although the mechanism by which cabozantinib suppresses radionuclide incorporation into foci undergoing bone remodeling remains unknown, that this phenomenon occurs in tumor-naïve models indicates that caution should be exercised in interpreting the clinical significance of this event.
Collapse
|
45
|
Ségaliny AI, Tellez-Gabriel M, Heymann MF, Heymann D. Receptor tyrosine kinases: Characterisation, mechanism of action and therapeutic interests for bone cancers. J Bone Oncol 2015; 4:1-12. [PMID: 26579483 PMCID: PMC4620971 DOI: 10.1016/j.jbo.2015.01.001] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 01/18/2015] [Indexed: 01/13/2023] Open
Abstract
Bone cancers are characterised by the development of tumour cells in bone sites, associated with a dysregulation of their environment. In the last two decades, numerous therapeutic strategies have been developed to target the cancer cells or tumour niche. As the crosstalk between these two entities is tightly controlled by the release of polypeptide mediators activating signalling pathways through several receptor tyrosine kinases (RTKs), RTK inhibitors have been designed. These inhibitors have shown exciting clinical impacts, such as imatinib mesylate, which has become a reference treatment for chronic myeloid leukaemia and gastrointestinal tumours. The present review gives an overview of the main molecular and functional characteristics of RTKs, and focuses on the clinical applications that are envisaged and already assessed for the treatment of bone sarcomas and bone metastases.
Collapse
Affiliation(s)
- Aude I Ségaliny
- INSERM, UMR 957, Equipe LIGUE Nationale Contre le Cancer 2012, Nantes 44035, France ; Université de Nantes, Nantes atlantique universités, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, Nantes, France
| | - Marta Tellez-Gabriel
- INSERM, UMR 957, Equipe LIGUE Nationale Contre le Cancer 2012, Nantes 44035, France ; Université de Nantes, Nantes atlantique universités, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, Nantes, France
| | - Marie-Françoise Heymann
- INSERM, UMR 957, Equipe LIGUE Nationale Contre le Cancer 2012, Nantes 44035, France ; Université de Nantes, Nantes atlantique universités, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, Nantes, France ; CHU de Nantes, France
| | - Dominique Heymann
- INSERM, UMR 957, Equipe LIGUE Nationale Contre le Cancer 2012, Nantes 44035, France ; Université de Nantes, Nantes atlantique universités, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, Nantes, France ; CHU de Nantes, France
| |
Collapse
|
46
|
Fay AP, Albiges L, Bellmunt J. Current role of cabozantinib in metastatic castration-resistant prostate cancer. Expert Rev Anticancer Ther 2015; 15:151-6. [PMID: 25586337 DOI: 10.1586/14737140.2015.1003047] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Over 80% of men with castration-resistant prostate cancer have bone metastases. This condition can dramatically impact quality of life and is associated with short-term survival. Consequently, the development of bone-targeted therapies is a relevant topic on prostate cancer management. Hepatocyte growth factor receptor and vascular endothelial growth factor signaling pathways have been identified to play a role in prostate cancer progression and bone metastasis and are potential targets for therapeutic intervention. Early-phase studies have shown encouraging responses in bone metastases and pain control with cabozantinib, a multi-tyrosine kinase inhibitor targeting hepatocyte growth factor receptor and vascular endothelial growth factor receptor. Despite striking responses seen in some patients, preliminary results from a pivotal Phase III study have failed to produce survival benefit. This review encompasses preclinical and clinical data of cabozantinib in metastatic castration-resistant prostate cancer highlighting future research options for this agent.
Collapse
Affiliation(s)
- André P Fay
- Dana-Farber Cancer Institute, Harvard Medical School 450 Brookline Avenue (DANA 1230), Boston, MA 02215, USA
| | | | | |
Collapse
|
47
|
|
48
|
Lee YH, Apolo AB, Agarwal PK, Bottaro DP. Characterization of HGF/Met Signaling in Cell Lines Derived From Urothelial Carcinoma of the Bladder. Cancers (Basel) 2014; 6:2313-29. [PMID: 25534569 PMCID: PMC4276968 DOI: 10.3390/cancers6042313] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 11/05/2014] [Accepted: 11/13/2014] [Indexed: 12/13/2022] Open
Abstract
There is mounting evidence of oncogenic hepatocyte growth factor (HGF)/Met signaling in urothelial carcinoma (UC) of the bladder. The effects of three kinase inhibitors, cabozantinib, crizotinib and EMD1214063, on HGF-driven signaling and cell growth, invasion and tumorigenicity were analyzed in cultured UC cell lines. SW780 xenograft growth in SCID and human HGF knock-in SCID (hHGF/SCID) mice treated with cabozantinib or vehicle, as well as tumor levels of Met and pMet, were also determined. Met content was robust in most UC-derived cell lines. Basal pMet content and effector activation state in quiescent cells were low, but significantly enhanced by added HGF, as were cell invasion, proliferation and anchorage independent growth. These HGF-driven effects were reversed by Met inhibitor treatment. Tumor xenograft growth was significantly higher in hHGF/SCID mice vs. SCID mice and significantly inhibited by cabozantinib, as was tumor phospho-Met content. These studies indicate the prevalence and functionality of the HGF/Met signaling pathway in UC cells, suggest that paracrine HGF may contribute to UC tumor growth and progression, and that support further preclinical investigation of Met inhibitors for the treatment of UC is warranted.
Collapse
Affiliation(s)
- Young H Lee
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Andrea B Apolo
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Piyush K Agarwal
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Donald P Bottaro
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
49
|
Cabanillas ME, Brose MS, Holland J, Ferguson KC, Sherman SI. A phase I study of cabozantinib (XL184) in patients with differentiated thyroid cancer. Thyroid 2014; 24:1508-14. [PMID: 25102375 PMCID: PMC4195402 DOI: 10.1089/thy.2014.0125] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Cabozantinib targets tyrosine kinases including MET, vascular endothelial growth factor (VEGF) receptor 2, and rearranged during transfection (RET). Differentiated thyroid cancer (DTC) is a tumor type that may be sensitive to cabozantinib. Therefore, we evaluated cabozantinib in a cohort of heavily pretreated patients with metastatic DTC. METHODS This single-arm open-label phase I trial assessed the safety, tolerability, and antitumor activity of cabozantinib in DTC patients taking part in a drug-drug interaction study. Adult patients with histologically confirmed metastatic or surgically unresectable DTC (including papillary, follicular, or Hürthle cell) were enrolled. Patients received daily oral dosing of 140 mg cabozantinib. Safety was assessed by evaluation of adverse events (AEs), vital signs, electrocardiograms, laboratory tests, and concomitant medications. Tumor response by magnetic resonance imaging or computed tomography scan was investigator assessed using Response Evaluation Criteria In Solid Tumors (RECIST) v1.0. RESULTS The study enrolled 15 patients who had failed standard radioactive iodine therapy. Patients had received a median of two prior systemic agents, and 11 patients (73%) had previously received at least one VEGF pathway inhibiting therapy. Common AEs included diarrhea, nausea, fatigue, and decreased appetite. Partial response was reported in eight patients (53%). Median progression-free survival and median overall survival were not reached. CONCLUSIONS Cabozantinib demonstrates a safety profile similar to other multitargeted VEGFR inhibitors in advanced DTC patients. The antitumor activity observed in this study warrants further investigation of cabozantinib in patients with advanced DTC.
Collapse
Affiliation(s)
- Maria E. Cabanillas
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Marcia S. Brose
- Department of Otorhinolaryngology–Head and Neck Surgery, University of Pennsylvania Perlman School of Medicine, Philadelphia, Pennsylvania
| | | | | | - Steven I. Sherman
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| |
Collapse
|
50
|
Song EK, Tai WM, Messersmith WA, Bagby S, Purkey A, Quackenbush KS, Pitts TM, Wang G, Blatchford P, Yahn R, Kaplan J, Tan AC, Atreya CE, Eckhardt G, Kelley RK, Venook A, Kwak EL, Ryan D, Arcaroli JJ. Potent antitumor activity of cabozantinib, a c-MET and VEGFR2 inhibitor, in a colorectal cancer patient-derived tumor explant model. Int J Cancer 2014; 136:1967-75. [PMID: 25242168 DOI: 10.1002/ijc.29225] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 08/11/2014] [Accepted: 09/03/2014] [Indexed: 12/17/2022]
Abstract
Antiangiogenic therapy is commonly used for the treatment of colorectal cancer (CRC). Although patients derive some clinical benefit, treatment resistance inevitably occurs. The MET signaling pathway has been proposed to be a major contributor of resistance to antiangiogenic therapy. MET is upregulated in response to vascular endothelial growth factor pathway inhibition and plays an essential role in tumorigenesis and progression of tumors. In this study, we set out to determine the efficacy of cabozantinib in a preclinical CRC patient-derived tumor xenograft model. We demonstrate potent inhibitory effects on tumor growth in 80% of tumors treated. The greatest antitumor effects were observed in tumors that possess a mutation in the PIK3CA gene. The underlying antitumor mechanisms of cabozantinib consisted of inhibition of angiogenesis and Akt activation and significantly decreased expression of genes involved in the PI3K pathway. These findings support further evaluation of cabozantinib in patients with CRC. PIK3CA mutation as a predictive biomarker of sensitivity is intriguing and warrants further elucidation. A clinical trial of cabozantinib in refractory metastatic CRC is being activated.
Collapse
Affiliation(s)
- Eun-Kee Song
- Division of Medical Oncology, University of Colorado Denver and University of Colorado Cancer Center, Aurora, CO; Department of Internal Medicine, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|