1
|
Voogd DWM, Lucassen MJJ, van der Noll R, Zielhuis SWJ, Boss D, Beijnen JH, Rosing H, Tibben M, Huitema ADR, Schellens JHM, Steeghs N. Phase I Study of Sorafenib Combined with Gemcitabine and Carboplatin in Patients with Advanced Solid Tumors. Oncol Ther 2025:10.1007/s40487-025-00340-8. [PMID: 40366624 DOI: 10.1007/s40487-025-00340-8] [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: 02/05/2025] [Accepted: 04/08/2025] [Indexed: 05/15/2025] Open
Abstract
INTRODUCTION A combination of targeted anticancer drugs with cytotoxic therapy can potentially overcome multidrug resistance. The multi-target kinase inhibitor sorafenib demonstrates synergistic activity when combined with chemotherapeutics in preclinical models. This phase I trial aimed to assess safety, tolerability, efficacy, and pharmacokinetics of sorafenib with gemcitabine and carboplatin. METHODS This single-center, open-label, dose-escalation and dose-expansion study included patients with advanced solid tumors considered for palliative treatment with gemcitabine and carboplatin. The maximum tolerated dose (MTD) was determined using a classic 3 + 3 dose-escalation design. Antitumor activity was evaluated every two treatment cycles. RESULTS In total, 45 patients received treatment. Of the patients, 49% (n = 22) were male, and median age was 58 years [range: 27-72 years]. After dose-escalation, sorafenib 400 mg once daily (q.d.) on days 1-21, gemcitabine 500 mg/m2 on day 1 and day 8 (D1D8), and carboplatin AUC3 on day 1 (D1) every 3 weeks (Q3W) were established as the MTD. Grade 4 treatment-related toxicities, all hematological, were seen in 22% of the patients. Frequently observed grade 3 adverse events were neutropenia (33%), thrombocytopenia (31%), leukopenia (16%), and fatigue (13%). Dose reductions were required in 33% of the patients across all dose levels. Disease control rate after 18 weeks was 50%. Median progression-free survival and overall survival were 5.4 months and 10.1 months, respectively. CONCLUSIONS A recommended phase 2 regimen of sorafenib 400 mg q.d. combined with gemcitabine 500 mg/m2 D1D8 and carboplatin AUC3 D1, Q3W showed a manageable toxicity profile. This combination could provide an effective treatment option for patients in whom other therapies have failed since antitumor activity was seen across heavily pretreated tumor types. Alternative dosing regimens should be studied to optimize the dosing schedule. TRIAL REGISTRATION EudraCT: 2007-004129-75.
Collapse
Affiliation(s)
- Daphne W M Voogd
- Department of Medical Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Merel J J Lucassen
- Department of Medical Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| | - Ruud van der Noll
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sybrand W J Zielhuis
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - David Boss
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Utrecht Institute of Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Hilde Rosing
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Matthijs Tibben
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jan H M Schellens
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Utrecht Institute of Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Neeltje Steeghs
- Department of Medical Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
2
|
Fu ZH, Zhao C, Wang Y, Zhang L, Wang L. Pharmacovigilance imbalance analysis of VEGFR-TKI-related taste and smell disorders. Sci Rep 2025; 15:3118. [PMID: 39856344 PMCID: PMC11760945 DOI: 10.1038/s41598-025-87678-1] [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: 07/23/2024] [Accepted: 01/21/2025] [Indexed: 01/27/2025] Open
Abstract
Taste and smell disorders (TSDs) can induce diminished interest in food, inadequate nutrient intake, and emotional irregularities, particularly among cancer patients. Previous research found that the main culprits of TSD development in cancer patients are cytotoxic drugs such as taxol, fluorouracil, cyclophosphamide, and anthracycline-based drugs. The advent of targeted drugs such as vascular endothelial growth factor receptor-tyrosine kinase inhibitors (VEGFR-TKIs) has significantly extended the survival time of cancer patients, and thus widely used in clinical practice. However, the association between the use of VEGFR-TKIs and the development of TSDs havs not been studied.The adverse event(AE) reports related to VEGFR-TKIs were downloaded from the FDA Adverse Event Reporting System (FAERS) database. Disproportionality analysis was conducted to assess the correlation between VEGFR-TKIs and TSDs. The Standardized Medical Dictionary for Regulatory Activities (MedDRA) Queries (SMQs) were used to analyze the AEs of TSDs. The study found a statistically significant correlation between the occurrence of TSDs and the use of VEGF-TKIs (cabozantinib, axitinib, pazopanib, sunitinib, nintedanib, and lenvatinib).However, the instructions for Nintedanib, Sorafenib and Lenvatinib were not mentioned. Capbottinib demonstrated the highest number of reports(1790 cases), also with the strongest association (ROR 95%CI-low = 16.51; PRR = 16.18; IC025 = 3.96) when analyzing the narrow SMQ of TSDs. Dysgeusia, taste disorder, and ageusia were the most commonly reported preferred terms (PTs) in VEGFR-TKI-related TSDs, accounting for more than 90% of the reported cases. Cabozantinib showed the highest number of reports and strongest correlation with ageusia, taste disorder, parosmia, and anosmia. The study found significant association between the reports of TSDs and the use of VEGFR-TKIs, indicating the monitoring of TSD development and appropriate management in clinical is necessary.
Collapse
Affiliation(s)
- Zhong-Hua Fu
- Department of pharmacy, School of Clinical Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Chenglong Zhao
- Department of pharmacy, School of Clinical Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Yaqin Wang
- Department of pharmacy, School of Clinical Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Lei Zhang
- Department of pharmacy, School of Clinical Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Lei Wang
- Department of pharmacy, School of Clinical Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China.
| |
Collapse
|
3
|
Yang L, Zhang Q, Xiong Y, Dang Z, Xiao H, Chen Q, Dai X, Zhang L, Zhu J, Wang D, Li M. A subset of VEGFR-TKIs activates AMPK in LKB1-mutant lung cancer. Cancer Sci 2022; 114:1651-1662. [PMID: 36459496 PMCID: PMC10067398 DOI: 10.1111/cas.15677] [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: 09/06/2022] [Revised: 11/10/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
The mutation of tumor suppressor gene liver kinase B1 (LKB1) has a prevalence of about 20% in non-small cell lung cancer (NSCLC). LKB1-mutant lung cancer is characterized by enhanced aggressiveness and immune escape and is associated with poor prognosis. Therefore, it is urgent to develop effective therapeutic methods for LKB1-mutant NSCLC. Recently, apatinib, a VEGFR-TKI, was found to significantly improve the outcome of LKB1-mutant NSCLC, but the mechanism is not completely clear. In this study, AMP-activated protein kinase (AMPK), the crucial downstream kinase of LKB1 was excavated as the potential target of apatinib. Biochemical experiments verified that apatinib is a direct AMPK activator. Moreover, clinically available VEGFR-TKIs were found to regulate AMPK differently: Apatinib and anlotinib can directly activate AMPK, while axitinib and sunitinib can directly inhibit AMPK. Activation of AMPK by apatinib leads to the phosphorylation of acetyl-CoA carboxylase (ACC) and inhibition of de novo fatty acid synthesis (FAsyn), which is upregulated in LKB1-null cancers. Moreover, the killing effect of apatinib was obviously enhanced under delipidated condition, and the combination of exogenous FA restriction with apatinib treatment can be a promising method for treating LKB1-mutant NSCLC. This study discovered AMPK as an important off-target of apatinib and elucidated different effects of this cluster of VEGFR-TKIs on AMPK. This finding can be the basis for the accurate and combined application of these drugs in clinic and highlights that the subset of VEGFR-TKIs including apatinib and anlotinib are potentially valuable in the treatment of LKB1-mutant NSCLC.
Collapse
Affiliation(s)
- Lujie Yang
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Qin Zhang
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Yanli Xiong
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhaoqian Dang
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - He Xiao
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Qian Chen
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiaoyan Dai
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Lei Zhang
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Jianwu Zhu
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Dong Wang
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Mengxia Li
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
4
|
Anderson TS, Wooster AL, Piersall SL, Okpalanwaka IF, Lowe DB. Disrupting cancer angiogenesis and immune checkpoint networks for improved tumor immunity. Semin Cancer Biol 2022; 86:981-996. [PMID: 35149179 PMCID: PMC9357867 DOI: 10.1016/j.semcancer.2022.02.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/11/2022] [Accepted: 02/05/2022] [Indexed: 01/27/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have advanced the field of cancer immunotherapy in patients by sustaining effector immune cell activity within the tumor microenvironment. However, the approach in general is still faced with issues related to ICI response duration/resistance, treatment eligibility, and safety, which indicates a need for further refinements. As immune checkpoint upregulation is inextricably linked to cancer-induced angiogenesis, newer clinical efforts have demonstrated the feasibility of disrupting both tumor-promoting networks to mediate enhanced immune-driven protection. This review focuses on such key evidence stipulating the necessity of co-applying ICI and anti-angiogenic strategies in cancer patients, with particular interest in highlighting newer engineered antibody approaches that may provide theoretically superior multi-pronged and safe therapeutic combinations.
Collapse
Affiliation(s)
- Trevor S Anderson
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Amanda L Wooster
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Savanna L Piersall
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Izuchukwu F Okpalanwaka
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Devin B Lowe
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States.
| |
Collapse
|
5
|
Angelousi A, Hayes AR, Chatzellis E, Kaltsas GA, Grossman AB. Metastatic medullary thyroid carcinoma: a new way forward. Endocr Relat Cancer 2022; 29:R85-R103. [PMID: 35521769 PMCID: PMC9175549 DOI: 10.1530/erc-21-0368] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/26/2022] [Indexed: 11/21/2022]
Abstract
Medullary thyroid carcinoma (MTC) is a rare malignancy comprising 1-2% of all thyroid cancers in the United States. Approximately 20% of cases are familial, secondary to a germline RET mutation, while the remaining 80% are sporadic and also harbour a somatic RET mutation in more than half of all cases. Up to 15-20% of patients will present with distant metastatic disease, and retrospective series report a 10-year survival of 10-40% from time of first metastasis. Historically, systemic therapies for metastatic MTC have been limited, and cytotoxic chemotherapy has demonstrated poor objective response rates. However, in the last decade, targeted therapies, particularly multitargeted tyrosine kinase inhibitors (TKIs), have demonstrated prolonged progression-free survival in advanced and progressive MTC. Both cabozantinib and vandetanib have been approved as first-line treatment options in many countries; nevertheless, their use is limited by high toxicity rates and dose reductions are often necessary. New generation TKIs, such as selpercatinib or pralsetinib, that exhibit selective activity against RET, have recently been approved as a second-line treatment option, and they exhibit a more favourable side-effect profile. Peptide receptor radionuclide therapy or immune checkpoint inhibitors may also constitute potential therapeutic options in specific clinical settings. In this review, we aim to present all current therapeutic options available for patients with progressive MTC, as well as new or as yet experimental treatments.
Collapse
Affiliation(s)
- Anna Angelousi
- Unit of Endocrinology, First Department of Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Athens, Greece
- Correspondence should be addressed to A Angelousi or A B Grossman: or
| | - Aimee R Hayes
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, Royal Free Hospital, London, UK
| | - Eleftherios Chatzellis
- Endocrinology Diabetes and Metabolism Department, 251 Hellenic Air Force and VA General Hospital, Athens, Greece
| | - Gregory A Kaltsas
- First Department of Propaedeutic Internal Medicine, Laiko Hospital, National & Kapodistrian University of Athens, Athens, Greece
| | - Ashley B Grossman
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, Royal Free Hospital, London, UK
- Green Templeton College, University of Oxford, Oxford, UK
- Centre for Endocrinology, Barts and the London School of Medicine, London, UK
- Correspondence should be addressed to A Angelousi or A B Grossman: or
| |
Collapse
|
6
|
Wang Y, Qiao X, Yang X, Yuan M, Xian S, Zhang L, Yang D, Liu S, Dai F, Tan Z, Cheng Y. The role of a drug-loaded poly (lactic co-glycolic acid) (PLGA) copolymer stent in the treatment of ovarian cancer. Cancer Biol Med 2021; 17:237-250. [PMID: 32296591 PMCID: PMC7142835 DOI: 10.20892/j.issn.2095-3941.2019.0169] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 09/11/2019] [Indexed: 12/12/2022] Open
Abstract
Objectives: Cisplatin (CDDP) is a widely used and effective basic chemotherapeutic drug for the treatment of a variety of tumors, including ovarian cancer. However, adverse side effects and acquired drug resistance are observed in the clinical application of CDDP. Identifying a mode of administration that can alleviate side effects and reduce drug resistance has become a promising strategy to solve this problem. Methods: In this study, 3D printing technology was used to prepare a CDDP-poly (lactic-co-glycolic acid) (CDDP-PLGA) polymer compound stent, and its physicochemical properties and cytotoxicity were evaluated both in vitro and in vivo. Results: The CDDP-PLGA stent had a significant effect on cell proliferation and apoptosis and clearly decreased the size of subcutaneous tumors in nude mice, whereas the systemic side effects were mild compared with those of intraperitoneal CDDP injection. Compared with the control group, CDDP-PLGA significantly increased the mRNA and protein levels of p-glycoprotein (P < 0.01; P < 0.01) and decreased vascular endothelial growth factor mRNA (P < 0.05) and protein levels (P < 0.01), however, CDDP-PLGA significantly decreased the mRNA and protein levels of p-glycoprotein (P < 0.01; P < 0.01) and vascular endothelial growth factor (P < 0.01; P < 0.01), which are associated with chemoresistance, in subcutaneous tumor tissue. Immunohistochemistry assay results revealed that, in the CDDP-PLGA group, the staining of the proliferation-related genes Ki67 and PCNA were lightly, and the apoptosis-related gene caspase-3 stained deeply. Conclusions: PLGA biomaterials loaded with CDDP, as compared with the same amount of free CDDP, showed good efficacy in terms of cytotoxicity, as evidenced by changes in apoptosis. Continuous local CDDP release can decrease the systemic side effects of this drug and the occurrence of drug resistance and angiogenesis, and improve the therapeutic effect. This new approach may be an effective strategy for the local treatment of epithelial ovarian cancer.
Collapse
Affiliation(s)
- Yanqing Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaoyin Qiao
- College of Biology, Hunan University, Changsha 410082, China
| | - Xiao Yang
- Department of Obstetrics and Gynecology, Renmin Hospital of Peking University, Beijing 100044, China
| | - Mengqin Yuan
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shu Xian
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Li Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Dongyong Yang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shiyi Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fangfang Dai
- Department of Obstetrics and Gynecology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Zhikai Tan
- College of Biology, Hunan University, Changsha 410082, China
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
7
|
Wang Q, Yan H, Wang Z, Li Z, Li D, Li Z, Wang K, Tian J, Zhao X. Construction of a novel bispecific fusion protein to enhance targeting for pancreatic cancer imaging. Biomaterials 2020; 255:120161. [PMID: 32505034 DOI: 10.1016/j.biomaterials.2020.120161] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 05/03/2020] [Accepted: 05/28/2020] [Indexed: 02/07/2023]
Abstract
Early detection and diagnosis are the most important endeavors for reducing associated morbidity and mortality of pancreatic ductal adenocarcinoma (PDAC). Developing molecular imaging probes that can specifically and effectively target cancer-associated biological pathways is one of the key points for sensitive and accurate diagnosis for PDAC. Herein, a small-sized, bispecific fusion protein constructed by genetic fusion of different binding domains of antibodies, termed Bi50, with enhanced targeting effect for PDAC is reported. Bi50 has excellent bispecific targeting for vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR) simultaneously in vitro and in vivo. Additionally, Bi50 shows increased intratumoral permeability and enrichment characteristics in the tumor than the control protein, which is constructed directly connecting two individual Fabs. Moreover, Bi50 can not only target areas rich in vasculature but also bind with affinity to tumor parenchymal cells, achieving "multilevel" targeting effect. Our work demonstrates that the bispecific fusion protein Bi50 has great potential as an efficient, targeted molecular imaging probe.
Collapse
Affiliation(s)
- Qian Wang
- Department of Diagnostic Imaging, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; CAS Key Laboratory of Molecular Imaging, Institute of Automation Chinese Academy of Sciences, Beijing, 100190, China
| | - Hao Yan
- Harvard Medical School and Wellman Center for Photomedicine, Massachusetts General Hospital, 50 Blossom Street, Boston, MA, 02114, USA
| | - Zihua Wang
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350108, Fujian Province, China
| | - Zhangfu Li
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Dan Li
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Zheng Li
- Yi-Chuang Institute of Biotechnology Industry, Beijing, 101111, China
| | - Kun Wang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation Chinese Academy of Sciences, Beijing, 100190, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation Chinese Academy of Sciences, Beijing, 100190, China.
| | - Xinming Zhao
- Department of Diagnostic Imaging, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
8
|
Borgatti A, Fieberg A, Winter AL, Stuebner K, Taras E, Todhunter D, Masyr A, Rendhal A, Vallera DA, Koopmeiners JS, Modiano JF. Impact of repeated cycles of EGF bispecific angiotoxin (eBAT) administered at a reduced interval from doxorubicin chemotherapy in dogs with splenic haemangiosarcoma. Vet Comp Oncol 2020; 18:664-674. [PMID: 32187827 DOI: 10.1111/vco.12590] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 12/13/2022]
Abstract
We previously reported that eBAT, an EGF-targeted angiotoxin, was safe and it improved the overall survival for dogs with splenic haemangiosarcoma when added to the standard of care in a single cycle of three administrations in the minimal residual disease setting. Our objective for the SRCBST-2 trial was to assess whether increased dosing through multiple cycles of eBAT would be well tolerated and would further enhance the benefits of eBAT. Eligibility was expanded to dogs with stage 3 haemangiosarcoma, provided that gross lesions could be surgically excised. The interval between eBAT and the start of chemotherapy was reduced, and the experimental therapy was expanded to three cycles, each administered at the biologically active dose (50 μg/kg) on a Monday/Wednesday/Friday schedule following splenectomy, and scheduled 1 week prior to the first, second and fifth doxorubicin chemotherapy. Twenty-five dogs were enrolled; six experienced acute hypotension with two requiring hospitalization. Self-limiting elevation of ALT was observed in one dog. A statistically significant survival benefit was not seen in this study in eBAT-treated dogs compared with a Contemporary comparison group of dogs with stages 1-3 haemangiosarcoma treated with standard of care alone. Our results indicate that repeated dosing cycles of eBAT starting 1 week prior to doxorubicin chemotherapy led to greater toxicity and reduced efficacy compared with a single cycle given between surgery and a delayed start of chemotherapy. Further work is needed to understand the precise mechanisms of action of eBAT in order to optimize its clinical benefits in the treatment of canine haemangiosarcoma and other tumours. IACUC Protocols 1110A06186 and 1507-32804A.
Collapse
Affiliation(s)
- Antonella Borgatti
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota, USA.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.,Clinical Investigation Center, College of Veterinary Medicine, St. Paul, Minnesota, USA
| | - Ann Fieberg
- Coordinating Center for Biometric Research, University of Minnesota, Minneapolis, Minnesota, USA
| | - Amber L Winter
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota, USA.,Clinical Investigation Center, College of Veterinary Medicine, St. Paul, Minnesota, USA
| | - Kathleen Stuebner
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota, USA.,Clinical Investigation Center, College of Veterinary Medicine, St. Paul, Minnesota, USA
| | - Elizabeth Taras
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.,Department of Radiation Oncology, School of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Deborah Todhunter
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.,Department of Radiation Oncology, School of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Alison Masyr
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Aaron Rendhal
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota, USA.,Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Daniel A Vallera
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.,Department of Radiation Oncology, School of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Joseph S Koopmeiners
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.,Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jaime F Modiano
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota, USA.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.,Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA.,Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
9
|
Lanera C, Minto C, Sharma A, Gregori D, Berchialla P, Baldi I. Extending PubMed searches to ClinicalTrials.gov through a machine learning approach for systematic reviews. J Clin Epidemiol 2018; 103:22-30. [PMID: 29981872 DOI: 10.1016/j.jclinepi.2018.06.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 06/19/2018] [Accepted: 06/29/2018] [Indexed: 12/23/2022]
Abstract
OBJECTIVES Despite their essential role in collecting and organizing published medical literature, indexed search engines are unable to cover all relevant knowledge. Hence, current literature recommends the inclusion of clinical trial registries in systematic reviews (SRs). This study aims to provide an automated approach to extend a search on PubMed to the ClinicalTrials.gov database, relying on text mining and machine learning techniques. STUDY DESIGN AND SETTING The procedure starts from a literature search on PubMed. Next, it considers the training of a classifier that can identify documents with a comparable word characterization in the ClinicalTrials.gov clinical trial repository. Fourteen SRs, covering a broad range of health conditions, are used as case studies for external validation. A cross-validated support-vector machine (SVM) model was used as the classifier. RESULTS The sensitivity was 100% in all SRs except one (87.5%), and the specificity ranged from 97.2% to 99.9%. The ability of the instrument to distinguish on-topic from off-topic articles ranged from an area under the receiver operator characteristic curve of 93.4% to 99.9%. CONCLUSION The proposed machine learning instrument has the potential to help researchers identify relevant studies in the SR process by reducing workload, without losing sensitivity and at a small price in terms of specificity.
Collapse
Affiliation(s)
- Corrado Lanera
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Via Loredan 18, Padova 35131, Italy
| | - Clara Minto
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Via Loredan 18, Padova 35131, Italy
| | - Abhinav Sharma
- Department of Biological Sciences and Bioengineering (BSBE), IIT, Kanpur, India
| | - Dario Gregori
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Via Loredan 18, Padova 35131, Italy
| | - Paola Berchialla
- Department of Clinical and Biological Sciences, University of Torino, Via Santena 5bis, Torino 10126, Italy
| | - Ileana Baldi
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Via Loredan 18, Padova 35131, Italy.
| |
Collapse
|
10
|
Bazaz R, Denning DW. Subacute Invasive Aspergillosis Associated With Sorafenib Therapy for Hepatocellular Carcinoma. Clin Infect Dis 2018; 67:156-157. [DOI: 10.1093/cid/ciy038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Rohit Bazaz
- National Aspergillosis Centre, Manchester University National Health Service Foundation Trust, United Kingdom
| | - David W Denning
- National Aspergillosis Centre, Manchester University National Health Service Foundation Trust, United Kingdom
- The University of Manchester and Manchester Academic Health Science Centre, United Kingdom
| |
Collapse
|
11
|
Borriello A, Caldarelli I, Bencivenga D, Stampone E, Perrotta S, Oliva A, Della Ragione F. Tyrosine kinase inhibitors and mesenchymal stromal cells: effects on self-renewal, commitment and functions. Oncotarget 2018; 8:5540-5565. [PMID: 27750212 PMCID: PMC5354929 DOI: 10.18632/oncotarget.12649] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/04/2016] [Indexed: 12/18/2022] Open
Abstract
The hope of selectively targeting cancer cells by therapy and eradicating definitively malignancies is based on the identification of pathways or metabolisms that clearly distinguish “normal” from “transformed” phenotypes. Some tyrosine kinase activities, specifically unregulated and potently activated in malignant cells, might represent important targets of therapy. Consequently, tyrosine kinase inhibitors (TKIs) might be thought as the “vanguard” of molecularly targeted therapy for human neoplasias. Imatinib and the successive generations of inhibitors of Bcr-Abl1 kinase, represent the major successful examples of TKI use in cancer treatment. Other tyrosine kinases have been selected as targets of therapy, but the efficacy of their inhibition, although evident, is less definite. Two major negative effects exist in this therapeutic strategy and are linked to the specificity of the drugs and to the role of the targeted kinase in non-malignant cells. In this review, we will discuss the data available on the TKIs effects on the metabolism and functions of mesenchymal stromal cells (MSCs). MSCs are widely distributed in human tissues and play key physiological roles; nevertheless, they might be responsible for important pathologies. At present, bone marrow (BM) MSCs have been studied in greater detail, for both embryological origins and functions. The available data are evocative of an unexpected degree of complexity and heterogeneity of BM-MSCs. It is conceivable that this grade of intricacy occurs also in MSCs of other organs. Therefore, in perspective, the negative effects of TKIs on MSCs might represent a critical problem in long-term cancer therapies based on such inhibitors.
Collapse
Affiliation(s)
- Adriana Borriello
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Ilaria Caldarelli
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Debora Bencivenga
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Emanuela Stampone
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Silverio Perrotta
- Department of Woman, Child and of General and Specialized Surgery, Second University of Naples, Naples, Italy
| | - Adriana Oliva
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Fulvio Della Ragione
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| |
Collapse
|
12
|
Totzeck M, Mincu RI, Mrotzek S, Schadendorf D, Rassaf T. Cardiovascular diseases in patients receiving small molecules with anti-vascular endothelial growth factor activity: A meta-analysis of approximately 29,000 cancer patients. Eur J Prev Cardiol 2018; 25:482-494. [DOI: 10.1177/2047487318755193] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Targeted therapy with tyrosine kinase inhibitors with anti-vascular endothelial growth factor activity improves survival of cancer patients. Cardiovascular complications are critical and it is unknown whether these require specific treatment strategies. We aimed to clarify the associated risk of cardiovascular adverse events in patients treated with tyrosine kinase inhibitors. Design The design of this study was a meta-analysis of randomised controlled trials. Methods We searched PubMed, Cochrane, EMBASE and Web of Science databases for randomised controlled trials published until January 2017 that assessed patients with different types of cancer treated with or without tyrosine kinase inhibitors in addition to standard chemotherapy. Results A total of 29,252 patients from 71 randomised controlled trials were included. Tyrosine kinase inhibitor treatment was associated with a higher cardiac ischaemia relative risk (relative risk = 1.69; 95% confidence interval: 1.12–2.57; p = 0.01), with the highest risks observed for sorafenib and patients with renal cancer. Risk of thrombocytopaenia (relative risk = 2.2; 95% confidence interval: 1.73–2.79; p < 0.001) was highest for regorafenib and patients with breast cancer. Left ventricular systolic dysfunction was increased after tyrosine kinase inhibitor therapy (relative risk = 2.53; 95% confidence interval:1.79 – 3.57; p < 0.001), with the highest risks reported for sunitinib and hepatocellular cancer. QT corrected interval prolongation (relative risk = 6.25; 95% confidence interval: 3.44–11.38; p < 0.001) and arterial hypertension (relative risk = 3.78; 95% confidence interval: 3.15-4.54; p < 0.001) were reported. The relative risks of arterial adverse events, cerebral ischaemia, venous adverse events and pulmonary embolism were similar across groups. Conclusion Tyrosine kinase inhibitors increase the risk of severe cardiovascular and particularly thrombotic adverse events. Specific treatment regimens when prescribing tyrosine kinase inhibitor therapies appear desirable.
Collapse
Affiliation(s)
- Matthias Totzeck
- Department of Cardiology and Vascular Medicine, University Hospital Essen, Germany
| | - Raluca-Ileana Mincu
- Department of Cardiology and Vascular Medicine, University Hospital Essen, Germany
| | - Simone Mrotzek
- Department of Cardiology and Vascular Medicine, University Hospital Essen, Germany
| | | | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, University Hospital Essen, Germany
| |
Collapse
|
13
|
VEGF pathway targeting agents, vessel normalization and tumor drug uptake: from bench to bedside. Oncotarget 2018; 7:21247-58. [PMID: 26789111 PMCID: PMC5008282 DOI: 10.18632/oncotarget.6918] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 12/05/2015] [Indexed: 12/19/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) pathway targeting agents have been combined with other anticancer drugs, leading to improved efficacy in carcinoma of the cervix, stomach, lung, colon and rectum, ovary, and breast. Vessel normalization induced by VEGF pathway targeting agents influences tumor drug uptake. Following bevacizumab treatment, preclinical and clinical studies have shown a decrease in tumor delivery of radiolabeled antibodies and two chemotherapeutic drugs. The decrease in vessel pore size during vessel normalization might explain the decrease in tumor drug uptake. Moreover, the addition of bevacizumab to cetuximab, or panitumumab in colorectal cancer patients or to trastuzumab in breast cancer patients, did not improve efficacy. However, combining bevacizumab with chemotherapy did increase efficacy in some cancer types. Novel biomarkers to select patients who may benefit from combination therapies, such as the effect of an angiogenesis inhibitor on tumor perfusion, requires innovative trial designs and large clinical trials. Small imaging studies with radiolabeled drugs could be used in the interphase to gain further insight into the interplay between VEGF targeted therapy, vessel normalization and tumor drug delivery.
Collapse
|
14
|
Priya SR, Dravid CS, Digumarti R, Dandekar M. Targeted Therapy for Medullary Thyroid Cancer: A Review. Front Oncol 2017; 7:238. [PMID: 29057215 PMCID: PMC5635342 DOI: 10.3389/fonc.2017.00238] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 09/19/2017] [Indexed: 12/16/2022] Open
Abstract
Medullary thyroid cancers (MTCs) constitute between 2 and 5% of all thyroid cancers. The 10-year overall survival (OS) rate of patients with localized disease is around 95% while that of patients with regional stage disease is about 75%. Only 20% of patients with distant metastases at diagnosis survive 10 years which is significantly lower than for differentiated thyroid cancers. Cases with regional metastases at presentation have high recurrence rates. Adjuvant external radiation confers local control but not improved OS. The management of residual, recurrent, or metastatic disease till a few years ago was re-surgery with local measures such as radiation. Chemotherapy was used with marginal benefit. The development of targeted therapy has brought in a major advantage in management of such patients. Two drugs—vandetanib and cabozantinib—have been approved for use in progressive or metastatic MTC. In addition, several drugs acting on other steps of the molecular pathway are being investigated with promising results. Targeted radionuclide therapy also provides an effective treatment option with good quality of life. This review covers the rationale of targeted therapy for MTC, present treatment options, drugs and methods under investigation, as well as an outline of the adverse effects and their management.
Collapse
Affiliation(s)
- S R Priya
- Head Neck Surgery, Homi Bhabha Cancer Hospital and Research Centre, Visakhapatnam, India.,Tata Memorial Centre, Mumbai, India
| | - Chandra Shekhar Dravid
- Head Neck Surgery, Homi Bhabha Cancer Hospital and Research Centre, Visakhapatnam, India.,Tata Memorial Centre, Mumbai, India
| | - Raghunadharao Digumarti
- Tata Memorial Centre, Mumbai, India.,Medical Oncology, Homi Bhabha Cancer Hospital and Research Centre, Visakhapatnam, India
| | | |
Collapse
|
15
|
Wouda RM, Hocker SE, Higginbotham ML. Safety evaluation of combination carboplatin and toceranib phosphate (Palladia) in tumour-bearing dogs: A phase I dose finding study. Vet Comp Oncol 2017; 16:E52-E60. [PMID: 28799187 DOI: 10.1111/vco.12332] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 05/03/2017] [Accepted: 05/12/2017] [Indexed: 01/06/2023]
Abstract
Combining conventional cytotoxic maximum tolerated dose (MTD) chemotherapy with low-dose metronomic and/or anti-angiogenic agents is a exciting area of oncologic research. The objective of this study was to establish the MTD, safety and adverse event (AE) profile of 1 such drug combination. This prospective phase I dose-finding clinical trial assumed an open-label 3 + 3 cohort design. Client-owned dogs with 1 or more cytologically and/or histologically confirmed and macroscopically measurable, naive or recurrent, malignant tumours, were enrolled. No preference for tumour histology, grade or stage was expressed. Toceranib was administered at a dose of 2.75 mg kg-1 by mouth (PO) every other day (EOD), and carboplatin administered intravenously (IV) every 21 days at a starting dose of 200 mg m-2 . A total of 25% dose escalation was proposed for carboplatin, to a maximum of 300 mg m-2 . AEs were graded according to the Veterinary Cooperative Oncology Group's common terminology criteria for AEs (VCOG-CTCAE). Grade 3 haematologic or gastrointestinal AEs were nominated dose-limiting. Response to therapy was evaluated according to the VCOG's revised RECIST criteria. Eleven dogs were enrolled. Tumour histologies included sinonasal carcinoma, osteosarcoma, thyroid carcinoma, melanoma and apocrine gland anal sac adenocarcinoma. MTDs of carboplatin and toceranib were identified as 200 mg m-2 IV every 21 days and approximately 2.75 mg kg-1 PO EOD, respectively. The dose-limiting toxicity was neutropenia. Two dogs experienced a partial response, and 6 maintained stable disease. Combination carboplatin and toceranib chemotherapy was well-tolerated. Clinical benefit was observed in most cases. This protocol warrants further investigation in phase II/III trials.
Collapse
Affiliation(s)
- R M Wouda
- Department of Clinical Sciences, Kansas State University, College of Veterinary Medicine, Manhattan, Kansas
| | - S E Hocker
- Department of Clinical Sciences, Kansas State University, College of Veterinary Medicine, Manhattan, Kansas
| | - M L Higginbotham
- Department of Clinical Sciences, Kansas State University, College of Veterinary Medicine, Manhattan, Kansas
| |
Collapse
|
16
|
Schaffrath J, Schmoll HJ, Voigt W, Müller LP, Müller-Tidow C, Mueller T. Efficacy of targeted drugs in germ cell cancer cell lines with differential cisplatin sensitivity. PLoS One 2017; 12:e0178930. [PMID: 28591197 PMCID: PMC5462387 DOI: 10.1371/journal.pone.0178930] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 05/22/2017] [Indexed: 12/25/2022] Open
Abstract
Testicular germ cell tumors (TGCTs) are the most common malignancies in men between the age of 15 and 35. Although cisplatin-based chemotherapy is highly effective in advanced disease, approximately 20% of patients have an unfavorable prognosis due to primary or acquired cisplatin resistance. For these patients, new therapeutic options are urgently needed. In numerous tumor entities, combinations of monoclonal antibodies or kinase inhibitors with chemotherapy exerted promising preclinical or clinical results, which have led to new treatment concepts. This prompted us to investigate the activity of different targeted agents alone or in combination with cisplatin in a panel of TGCT cell lines.
Collapse
Affiliation(s)
- Judith Schaffrath
- Department of Internal Medicine IV, Oncology/Hematology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Hans-Joachim Schmoll
- Workgroup Clinical Studies in Oncology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Wieland Voigt
- Medical Innovations and Management, Innovation in Oncology, Steinbeis University, Berlin, Germany
| | - Lutz P. Müller
- Department of Internal Medicine IV, Oncology/Hematology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Mueller
- Department of Internal Medicine IV, Oncology/Hematology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- * E-mail:
| |
Collapse
|
17
|
Borgatti A, Koopmeiners JS, Sarver AL, Winter AL, Stuebner K, Todhunter D, Rizzardi AE, Henriksen JC, Schmechel S, Forster CL, Kim JH, Froelich J, Walz J, Henson MS, Breen M, Lindblad-Toh K, Oh F, Pilbeam K, Modiano JF, Vallera DA. Safe and Effective Sarcoma Therapy through Bispecific Targeting of EGFR and uPAR. Mol Cancer Ther 2017; 16:956-965. [PMID: 28193671 PMCID: PMC5418099 DOI: 10.1158/1535-7163.mct-16-0637] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 01/19/2017] [Accepted: 01/23/2017] [Indexed: 01/12/2023]
Abstract
Sarcomas differ from carcinomas in their mesenchymal origin. Therapeutic advancements have come slowly, so alternative drugs and models are urgently needed. These studies report a new drug for sarcomas that simultaneously targets both tumor and tumor neovasculature. eBAT is a bispecific angiotoxin consisting of truncated, deimmunized Pseudomonas exotoxin fused to EGF and the amino terminal fragment of urokinase. Here, we study the drug in an in vivo "ontarget" companion dog trial as eBAT effectively kills canine hemangiosarcoma and human sarcoma cells in vitro We reasoned the model has value due to the common occurrence of spontaneous sarcomas in dogs and a limited lifespan allowing for rapid accrual and data collection. Splenectomized dogs with minimal residual disease were given one cycle of eBAT followed by adjuvant doxorubicin in an adaptive dose-finding, phase I-II study of 23 dogs with spontaneous, stage I-II, splenic hemangiosarcoma. eBAT improved 6-month survival from <40% in a comparison population to approximately 70% in dogs treated at a biologically active dose (50 μg/kg). Six dogs were long-term survivors, living >450 days. eBAT abated expected toxicity associated with EGFR targeting, a finding supported by mouse studies. Urokinase plasminogen activator receptor and EGFR are targets for human sarcomas, so thorough evaluation is crucial for validation of the dog model. Thus, we validated these markers for human sarcoma targeting in the study of 212 human and 97 canine sarcoma samples. Our results support further translation of eBAT for human patients with sarcomas and perhaps other EGFR-expressing malignancies. Mol Cancer Ther; 16(5); 956-65. ©2017 AACR.
Collapse
Affiliation(s)
- Antonella Borgatti
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota.
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Joseph S Koopmeiners
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Aaron L Sarver
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Amber L Winter
- Clinical Investigation Center, College of Veterinary Medicine, St. Paul, Minnesota
| | - Kathleen Stuebner
- Clinical Investigation Center, College of Veterinary Medicine, St. Paul, Minnesota
| | - Deborah Todhunter
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Radiation Oncology, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Anthony E Rizzardi
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Jonathan C Henriksen
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Stephen Schmechel
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Colleen L Forster
- BioNet Histology Research Laboratory, Academic Health Center, University of Minnesota, Minneapolis, Minnesota
| | - Jong-Hyuk Kim
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Jerry Froelich
- Department of Radiology, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Jillian Walz
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
| | - Michael S Henson
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Matthew Breen
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, and Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina
- Cancer Genetics Program, University of North Carolina Lineberger Comprehensive Cancer Center, Raleigh, North Carolina
| | - Kerstin Lindblad-Toh
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Felix Oh
- Department of Radiation Oncology, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Kristy Pilbeam
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Jaime F Modiano
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota
| | - Daniel A Vallera
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Radiation Oncology, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
18
|
Kakizawa N, Suzuki K, Fukui T, Takayama Y, Ichida K, Muto Y, Hasegawa F, Watanabe F, Kikugawa R, Tsujinaka S, Futsuhara K, Miyakura Y, Noda H, Rikiyama T. Clinical and molecular assessment of regorafenib monotherapy. Oncol Rep 2017; 37:2506-2512. [PMID: 28259999 DOI: 10.3892/or.2017.5456] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 01/30/2017] [Indexed: 11/05/2022] Open
Abstract
Regorafenib has shown survival benefits in metastatic colorectal cancer patients who were exacerbated after all standard therapies. Some patients, however, exhibit severe adverse events (AEs) resulting in treatment discontinuation. Therefore, the selection of patients likely to benefit from regorafenib is crucial. Twenty patients were treated with regorafenib for metastatic colorectal cancer; 122 plasma samples were taken from 16 of these patients for monitoring of circulating tumor DNA (ctDNA) in the blood. The treatment response, AEs, overall survival (OS), progression-free survival (PFS) and tumor morphologic changes on CT images were evaluated. KRAS mutant ctDNA was determined using digital PCR. Median PFS and OS were 2.5 and 5.9 months, respectively. Treatment was discontinued because of disease progression (PD) in 10 patients, and AEs in another 10 patients. AEs included hyperbilirubinemia, severe fatigue and skin rash. Hyperbilirubinemia was seen in two patients with multiple bilateral liver metastases, and severe fatigue in another 2 patients with poor performance status (PS). These severe AEs resulted in treatment discontinuation. Ten patients had a median PFS of 2.1 months with AE related discontinuation; PD occurred at 3.5 months (p=0.00334). Four patients exhibited a morphologic response, achieving better PFS times of 3.5, 5.3, 5.6 and 14.2 months. Emergence of the KRAS mutation in ctDNA was observed during anti-EGFR antibody treatment in 3 patients among 11 with KRAS wild-type tumors; it was detectable in the blood prior to radiographic detection of PD. Moreover, the KRAS mutation declined in two patients during regorafenib monotherapy. These patients were re-challenged with anti-EGFR antibody. Patients with extensive multiple liver metastases or poor PS are unlikely to benefit from regorafenib. Patients with a morphologic response will probably benefit from regorafenib with adequate management of other AEs. KRAS monitoring in ctDNA could be useful regarding treatment response and in determining treatment strategy.
Collapse
Affiliation(s)
- Nao Kakizawa
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Koichi Suzuki
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Taro Fukui
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Yuji Takayama
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Kosuke Ichida
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Yuta Muto
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Fumi Hasegawa
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Fumiaki Watanabe
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Rina Kikugawa
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Shingo Tsujinaka
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Kazushige Futsuhara
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Yasuyuki Miyakura
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Hiroshi Noda
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| | - Toshiki Rikiyama
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Omiya-ku, Saitama 330-8503, Japan
| |
Collapse
|
19
|
Kamel MK, Nasar A, Stiles BM, Altorki NK, Port JL. Video-Assisted Thoracoscopic Lobectomy Is the Preferred Approach Following Induction Chemotherapy. J Laparoendosc Adv Surg Tech A 2016; 27:495-500. [PMID: 27996367 DOI: 10.1089/lap.2016.0540] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE A video-assisted thoracoscopic surgical (VATS) resection, after induction chemotherapy, has long been considered a relative contraindication. We report our experience with VATS lobectomy after induction chemotherapy for patients with nonsmall cell lung cancer (NSCLC), with propensity-matched group of patients, who underwent an open approach, to determine safety and oncological outcome. METHODS A retrospective review of a prospective database (2002-2014) was performed to identify patients undergoing potentially curative lobectomy for NSCLC after induction therapy. Propensity score matching (age, gender, and clinical stage) was performed (1:2) to obtain a balanced cohort of patients undergoing VATS resection and thoracotomy. RESULTS A total of 285 patients underwent lobectomy after induction therapy, 114 were propensity matched (VATS, n = 40, thoracotomy, n = 74). There were no differences in the clinicopathological factors or type of induction therapy (conventional versus targeted) between VATS and thoracotomy groups. Similarly, no differences were found in the number of lymph nodes resected (12 versus 15, P = .94), the number of stations sampled (4 for each, P = .68), or in the rate of R0 resection (95% versus 96%, P = .81) between VATS and thoracotomy groups. Five VATS cases were converted to an open approach because of adhesions. VATS resection was associated with less estimated blood loss (EBL), shorter length of stay (LOS), and a trend toward fewer postoperative complications. There was no difference in 5 years disease-free survival (DFS) between VATS and thoracotomy groups (73% versus 48%, P = .09). Similarly, for patients who presented with cN2, there were no differences between thoracotomy and VATS groups in DFS (P = .37). On multi-variable analysis (MVA), only the clinical N1/2 status [Hazard ratio (HR): 4.86, P < .001] independently predicted poor DFS. CONCLUSIONS A VATS lobectomy is a feasible, safe, and oncologically sound approach after induction therapy for NSCLC. When compared with thoracotomy, VATS lobectomy is associated with lower EBL, shorter LOS, and a trend toward fewer postoperative complications.
Collapse
Affiliation(s)
- Mohamed K Kamel
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York Presbyterian Hospital , New York, New York
| | - Abu Nasar
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York Presbyterian Hospital , New York, New York
| | - Brendon M Stiles
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York Presbyterian Hospital , New York, New York
| | - Nasser K Altorki
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York Presbyterian Hospital , New York, New York
| | - Jeffrey L Port
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York Presbyterian Hospital , New York, New York
| |
Collapse
|
20
|
Targeting the angio-proteostasis network: Combining the forces against cancer. Pharmacol Ther 2016; 167:1-12. [DOI: 10.1016/j.pharmthera.2016.07.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/14/2016] [Indexed: 01/24/2023]
|
21
|
Koňariková K, Perdikaris GA, Gbelcova H, Andrezálová L, Švéda M, Ruml T, Laubertová L, Žitňanová I. Effect of Schiff base Cu(II) complexes on signaling pathways in HT-29 cells. Mol Med Rep 2016; 14:4436-4444. [PMID: 27633628 DOI: 10.3892/mmr.2016.5739] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 07/12/2016] [Indexed: 11/05/2022] Open
Abstract
Schiff base copper (II) complexes are known for their anticancer, antifungal, antiviral and anti‑inflammatory activities. The aim of the current study was to investigate biological effects of Schiff base Cu (II) complexes (0.001‑100 µmol/l)‑[Cu2(sal‑D, L‑glu)2(isoquinoline)2]·2C2H5OH (1), [Cu(sal‑5‑met‑L‑glu)(H2O)].H2O (2), [Cu(ethanol)2(imidazole)4][Cu2(sal‑D, L-glu)2(imidazole)2] (3), [Cu(sal‑D,L‑glu)(2‑methylimidazole)] (4) on the human colon carcinoma cells HT‑29, the mouse noncancerous cell line NIH‑3T3 and the human noncancerous fibroblast cell line VH10. The results suggested that Cu (II) complexes exhibit cytotoxic effects against the HT‑29 cell line, while complexes 3 and 4 were the most effective. Subsequent to 72 h of incubation, apoptosis was observed in the HT‑29 cells induced by Cu (II) complexes 1 (0.1, 1, 10 and 50 µmol/l), 2 (1, 10, 50 and 100 µmol/l), 3 (0.01, 1, 10 and 50 µmol/l) and 4 (0.01, 0.1, 1 and 10 µmol/l). The apoptotic pathways activated by the Cu (II) complexes were identified. The results indicated that complexes 2, 3 and 4 were able to induce the mitochondria‑dependent pathway of apoptosis in HT‑29 cells, while complex 1 was obsered to activate the extrinsic pathway of apoptosis. The levels of the anti‑apoptotic protein Bcl‑2 were reduced and those of the pro‑apoptotic protein Bax increased following treatment with complexes 2, 3 and 4. Complex 1 had no effect on Bax protein expression. Complexes 2 and 3 induced elevation of cytochrome c (cyt c), while complex 4 induced a time‑dependent elevation of cyt c levels. No cyt c was detected in HT‑29 cells exposed to complex 1, suggesting that Cu (II) complexes activated the extrinsic pathway of apoptosis. The results from the current study in addition to previous studies suggest that Schiff base Cu (II) complexes have potential as novel anticancer drugs.
Collapse
Affiliation(s)
- Katarína Koňariková
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University, 813 72 Bratislava, Slovak Republic
| | - Georgios A Perdikaris
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University, 813 72 Bratislava, Slovak Republic
| | - Helena Gbelcova
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, 813 72 Bratislava, Slovak Republic
| | - Lucia Andrezálová
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University, 813 72 Bratislava, Slovak Republic
| | - Martin Švéda
- Department of Biochemistry and Microbiology, Institute of Chemical Technology, Faculty of Food Biochemical Technology, 166 28 Prague, Czech Republic
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, Institute of Chemical Technology, Faculty of Food Biochemical Technology, 166 28 Prague, Czech Republic
| | - Lucia Laubertová
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University, 813 72 Bratislava, Slovak Republic
| | - Ingrid Žitňanová
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University, 813 72 Bratislava, Slovak Republic
| |
Collapse
|
22
|
A Phase I, Dose-Escalation Trial of Pazopanib in Combination with Cisplatin in Patients with Advanced Solid Tumors: A UNICANCER Study. Oncol Ther 2016; 4:211-223. [PMID: 28261651 PMCID: PMC5315079 DOI: 10.1007/s40487-016-0027-x] [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: 05/25/2016] [Indexed: 02/02/2023] Open
Abstract
Introduction To determine the feasibility, maximum-tolerated dose (MTD), and dose-limiting toxicities (DLT) of pazopanib in combination with cisplatin. Methods Patients with advanced malignancies were included in a 3 + 3 dose-escalation phase I study. Pazopanib administration started 8 days before the first infusion of cisplatin; some patients were treated according to a reverse sequence (cisplatin first). Five dose levels (DLs) were planned. MTD was based on DLT observed during cycles 1 and 2. Results Thirty-five patients were enrolled. The MTD was reached at the first DL, (pazopanib 400 mg daily + cisplatin 75 mg/m2 every 21 days). Main DLTs were pulmonary embolism, neutropenia, thrombocytopenia, and elevation of liver enzymes. Overall, most common adverse events were anemia (83%), fatigue (80%), thrombocytopenia (80%), neutropenia (73%), hypertension (59%), neurotoxicity (56%), and anorexia (53%). Sixteen patients (46%) discontinued the study due to toxicity. One patient (sarcoma) had a complete response, and three patients (one with breast cancer and two with ovarian cancers) had a partial response. Pharmacokinetic (PK) analyses showed interactions with aprepitant, resulting in increased exposure to pazopanib, which might explain partly the poor tolerance of the combination. Conclusion Cisplatin and pazopanib could not be administered at their single agent full doses, partly due to a PK interaction between pazopanib and aprepitant. Funding This work was funded by GlaxoSmithKline and by the charity Ligue Nationale de Lutte Contre le Cancer. Trial registered ClinicalTrials.gov identifier, NCT01165385.
Collapse
|
23
|
Update on Cardiovascular Safety of Tyrosine Kinase Inhibitors: With a Special Focus on QT Interval, Left Ventricular Dysfunction and Overall Risk/Benefit. Drug Saf 2016; 38:693-710. [PMID: 26008987 DOI: 10.1007/s40264-015-0300-1] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
We previously reviewed the cardiovascular safety of 16 tyrosine kinase inhibitors (TKIs), approved for use in oncology as of 30 September 2012. Since then, the indications for some of them have been widened and an additional nine TKIs have also been approved as of 30 April 2015. Eight of these nine are indicated for use in oncology and one (nintedanib) for idiopathic pulmonary fibrosis. This report is an update on the cardiovascular safety of those 16 TKIs, including the post-marketing data concerning their pro-arrhythmic effects, and reviews the cardiovascular safety of the nine new TKIs approved since (afatinib, cabozantinib, ceritinib, dabrafenib, ibrutinib, lenvatinib, nintedanib, ponatinib, and trametinib). As before, we focus on specific aspects of cardiovascular safety, namely their potential to induce QT interval prolongation, left ventricular (LV) dysfunction and hypertension but now also summarise the risks of arterial thromboembolic events (ATEs) associated with these agents. Of the newer TKIs, cabozantinib and ceritinib have been shown to induce a mild to moderate degree of QTc interval prolongation while cardiac dysfunction has been reported with the use of afatinib, dabrafenib, lenvatinib, ponatinib and trametinib. The label for axitinib was revised to include a new association with cardiac dysfunction. Hypertension is associated with cabozantinib, lenvatinib, nintedanib, ponatinib and trametinib. Ponatinib, within 10 months of its approval in December 2012, required voluntary (temporary) suspension of its marketing until significant safety revisions (restricted indication, additional warnings and precautions about the risk of arterial occlusion and thromboembolic events and amended dose) were made to its label. Compared with the previous 16 TKIs, more of the recently introduced TKIs are associated with the risk of LV dysfunction, and fewer with QT prolongation. Available data on morbidity and mortality associated with TKIs, together with post-marketing experience with lapatinib and ponatinib, emphasise the need for effective pharmacovigilance and ongoing re-assessment of their risk/benefit after approval of these novel agents. If not adequately managed, these cardiovascular effects significantly decrease the quality of life and increase the morbidity and mortality in a population already at high risk. Evidence accumulated over the last decade suggests that their clinical benefit, although worthwhile, is modest and extends only to progression-free survival and complete response without any effect on overall survival. During uncontrolled use in routine clinical practice, their risk/benefit is likely to be inferior to that perceived from highly controlled clinical trials.
Collapse
|
24
|
Reinwald M, Boch T, Hofmann WK, Buchheidt D. Risk of Infectious Complications in Hemato-Oncological Patients Treated with Kinase Inhibitors. Biomark Insights 2016; 10:55-68. [PMID: 27127405 PMCID: PMC4841329 DOI: 10.4137/bmi.s22430] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 09/13/2015] [Accepted: 09/15/2015] [Indexed: 12/30/2022] Open
Abstract
Infectious complications are a major cause of morbidity and mortality in patients with hemato-oncological diseases. Although disease-related immunosuppression represents one factor, aggressive treatment regimens, such as chemotherapy, stem cell transplantation, or antibody treatment, account for a large proportion of infectious side effects. With the advent of targeted therapies affecting specific kinases in malignant diseases, the outcome of patients has further improved. Nonetheless, dependent on the specific pathway targeted or off-target activity of the kinase inhibitor, therapy-associated infectious complications may occur. We review the most common and approved kinase inhibitors targeting a variety of hemato-oncological malignancies for their immunosuppressive potential and evaluate their risk of infectious side effects based on preclinical evidence and clinical data in order to raise awareness of the potential risks involved.
Collapse
Affiliation(s)
- Mark Reinwald
- Department of Hematology and Oncology, Mannheim University Hospital, University of Heidelberg, Mannheim, Germany
| | - Tobias Boch
- Department of Hematology and Oncology, Mannheim University Hospital, University of Heidelberg, Mannheim, Germany
| | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, Mannheim University Hospital, University of Heidelberg, Mannheim, Germany
| | - Dieter Buchheidt
- Department of Hematology and Oncology, Mannheim University Hospital, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
25
|
Straka C, Ying J, Kong FM, Willey CD, Kaminski J, Kim DWN. Review of evolving etiologies, implications and treatment strategies for the superior vena cava syndrome. SPRINGERPLUS 2016; 5:229. [PMID: 27026923 PMCID: PMC4771672 DOI: 10.1186/s40064-016-1900-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 02/17/2016] [Indexed: 12/25/2022]
Abstract
Superior vena cava syndrome (SVCS) is a relatively common sequela of mediastinal malignancies and may cause significant patient distress. SVCS is a medical emergency if associated with laryngeal or cerebral edema. The etiologies and management of SVCS have evolved over time. Non-malignant SVCS is typically caused by infectious etiologies or by thrombus in the superior vena cava and can be managed with antibiotics or anti-coagulation therapy, respectively. Radiation therapy (RT) has long been a mainstay of treatment of malignant SVCS. Chemotherapy has also been used to manage SVCS. In the past 20 years, percutaneous stenting of the superior vena cava has emerged as a viable option for SVCS symptom palliation. RT and chemotherapy are still the only modalities that can provide curative treatment for underlying malignant etiologies of SVCS. The first experiences with treating SVCS with RT were reported in the 1970’s, and several advances in RT delivery have subsequently occurred. Hypo-fractionated RT has the potential to be a more convenient therapy for patients and may provide equal or superior control of underlying malignancies. RT may be combined with stenting and/or chemotherapy to provide both immediate symptom palliation and long-term disease control. Clinicians should tailor therapy on a case-by-case basis. Multi-disciplinary care will maximize treatment expediency and efficacy.
Collapse
Affiliation(s)
- Christopher Straka
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, 5801 Forest Park Rd, Dallas, TX 75390 USA
| | - James Ying
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, 5801 Forest Park Rd, Dallas, TX 75390 USA
| | - Feng-Ming Kong
- Department of Radiation Oncology, GRU Cancer Center and Medical College of Georgia, Augusta, GA USA
| | - Christopher D Willey
- Department of Radiation Oncology, The University of Alabama Birmingham, Birmingham, AL USA
| | - Joseph Kaminski
- Dattoli Cancer Center, 2803 Fruitville Rd, Sarasota, FL 34237 USA
| | - D W Nathan Kim
- Department of Radiation Oncology, Texas Oncology, 1700 W. Highway 6, Waco, TX 76712 USA
| |
Collapse
|
26
|
Chang K, Karnad A, Zhao S, Freeman JW. Roles of c-Met and RON kinases in tumor progression and their potential as therapeutic targets. Oncotarget 2016; 6:3507-18. [PMID: 25784650 PMCID: PMC4414132 DOI: 10.18632/oncotarget.3420] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 01/21/2015] [Indexed: 02/06/2023] Open
Abstract
c-Met and receptor originated from nantes (RON) are structurally related transmembrane phosphotyrosine kinase receptors. c-Met and RON show increased expression or activity in a variety of tumors leading to tumor progression and may play a role in acquired resistance to therapy. Although often co-expressed, the distinct functional roles of c-Met and RON are not fully understood. c-Met and RON form both activated homodimers and heterodimers with themselves and other families of phosphotyrosine kinase receptors. Inhibitors for c-Met and RON including small molecular weigh kinase inhibitors and neutralizing antibodies are in pre-clinical investigation and clinical trials. Several of the tyrosine kinase inhibitors have activity against both c-Met and RON kinases whereas the antibodies generally are target specific. As with many targeted agents used to treat solid tumors, it is likely that c-Met/RON inhibitors will have greater benefit when used in combination with chemotherapy or other targeted agents. A careful analysis of c-Met/RON expression or activity and a better elucidation of how they influence cell signaling will be useful in predicting which tumors respond best to these inhibitors as well as determining which agents can be used with these inhibitors for combined therapy.
Collapse
Affiliation(s)
- Katherine Chang
- Department of Medicine, Division of Medical Oncology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Cancer Therapy and Research Center, Experimental and Developmental Therapeutics Program, San Antonio, TX, USA
| | - Anand Karnad
- Department of Medicine, Division of Medical Oncology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Cancer Therapy and Research Center, Experimental and Developmental Therapeutics Program, San Antonio, TX, USA
| | - Shujie Zhao
- Department of Medicine, Division of Medical Oncology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - James W Freeman
- Department of Medicine, Division of Medical Oncology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Cancer Therapy and Research Center, Experimental and Developmental Therapeutics Program, San Antonio, TX, USA.,Research and Development, Audie Murphy Veterans Administration Hospital, San Antonio, TX, USA
| |
Collapse
|
27
|
Gharwan H, Groninger H. Kinase inhibitors and monoclonal antibodies in oncology: clinical implications. Nat Rev Clin Oncol 2015; 13:209-27. [PMID: 26718105 DOI: 10.1038/nrclinonc.2015.213] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Molecularly targeted cancer therapies, such as small-molecule kinase inhibitors and monoclonal antibodies, constitute a rapidly growing and an important part of the oncology armamentarium. Unlike conventional (cytotoxic) chemotherapeutics, targeted therapies were designed to disrupt cancer cell pathogenesis at specific biological points essential for the development and progression of the tumour. These agents were developed to disrupt specific targets with the aim of minimizing treatment burden compared with conventional chemotherapy. Nevertheless the increasingly common use of targeted therapies has revealed some unanticipated, often clinically significant toxic effects, as well as compromising effective palliative and end-of-life management approaches. Although patients and clinicians welcome improvements in cancer prognosis, these changes can also impact patient quality-of-life. Therefore, as demand for oncology expertise increases, physicians need to apprise themselves of targeted therapies and their clinical implications, including drug-specific side effects, impact on quality of life, and cost issues, especially in relation to end-of-life care. This Review provides a useful summary and guide for professionals treating patients with malignant diseases.
Collapse
Affiliation(s)
- Helen Gharwan
- Medical Oncology, National Cancer Institute, National Institutes of Health, 10 Center Drive, Building 10, Room 12N226, Bethesda, Maryland 20892-1906, USA
| | - Hunter Groninger
- Section of Palliative Care, Department of Medicine, MedStar Washington Hospital Center, 110 Irving Street NW, Room 2A-68, Washington, District of Columbia 20008, USA
| |
Collapse
|
28
|
Li Y, Liang XY, Yue YQ, Sheng L, Liu JK, Wang ZY, Chen G. Does the addition of drugs targeting the vascular endothelial growth factor pathway to first-line chemotherapy increase complete response? A meta-analysis of randomized clinical trials. Tumour Biol 2015; 37:6297-306. [PMID: 26619847 DOI: 10.1007/s13277-015-4493-9] [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: 10/25/2015] [Accepted: 11/23/2015] [Indexed: 10/22/2022] Open
Abstract
Drugs targeting the vascular endothelial growth factor (VEGF) and its receptor (VEGFR) signaling (anti-VEGF/VEGFR drugs) are the most validated anti-angiogenic strategies for cancer treatment. Complete response (CR) is a rare event in cancer patients receiving chemotherapy. A meta-analysis was conducted to determine whether adding anti-VEGF/VEGFR drugs to chemotherapy can further increase the chance of CR in the first-line therapy. Relevant databases were systematically searched for the period 2000-2015. Eligible studies were selected according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. The incidence, relative risk (RR), and 95 % confidence intervals (CIs) were calculated using random-effects or fixed-effects models based on the heterogeneity of selected studies. A total of 12,453 patients from 28 randomized controlled trials were included. The overall incidence of CR in patients treated with anti-VEGF/VEGFR drugs plus chemotherapy was 1.5 % (95 % CI, 1.0-2.0 %) compared to 1.1 % (95 % CI, 0.7-1.4 %) in the chemotherapy-alone arm. Adding anti-VEGF/VEGFR drugs was associated with significant improvement of CR (RR, 1.52, 95 % CI, 1.18-1.95, P = 0.001). When stratified by drug type, adding VEGFR tyrosin kinase inhibitors (TKIs) did not increase the chance of CR (RR, 0.87, 95 % CI, 0.51-1.49; P = 0.614). The addition of bevacizumab with 7.5 mg/kg every 3 weeks, but not 15 mg/kg every 3 weeks, significantly improves the CR (7.5 mg, RR, 2.43, 95 % CI, 1.64-3.60, P = 0.000; 15 mg, RR, 1.07, 95 % CI, 0.63-1.81, P = 0.799). In subgroup analysis, a significant improvement of CR by the addition of anti-VEGF/VEGFR drugs was observed in patients with colorectal cancer (RR, 2.10, 95 % CI 1.21-3.63, P = 0.008), ovarian cancer (RR, 3.07; 95 % CI, 1.68-5.62, P = 0.000), and patients who are treated with platinum-based regimens (RR, 1.78, 95 % CI, 1.23-2.59, P = 0.002). Low-dose bevacizumab, rather than VEGFR TKIs or high-dose bevacizumab, can increase the chance of CR in patients receiving chemotherapy.
Collapse
Affiliation(s)
- Yan Li
- Department of Urology, Jinshan Hospital, Fudan University, 1508 Longhang Road, Shanghai, 201500, China
| | - Xin-Yue Liang
- Institute of Clinical Pharmacology, Qilu Hospital, Shandong University, Shandong, China
| | - Yi-Qi Yue
- Department of Gynecology, Xuhui District Central Hospital, Shanghai, China
| | - Lei Sheng
- Centre for Personalised Cancer Medicine, School of Medicine, The University of Adelaide, Adelaide, Australia
| | - Ji-Kai Liu
- Department of Urology, Jinshan Hospital, Fudan University, 1508 Longhang Road, Shanghai, 201500, China
| | - Zhan-Yu Wang
- Department of Urology, Jinshan Hospital, Fudan University, 1508 Longhang Road, Shanghai, 201500, China
| | - Gang Chen
- Department of Urology, Jinshan Hospital, Fudan University, 1508 Longhang Road, Shanghai, 201500, China.
| |
Collapse
|
29
|
Liao HW, Hsu JM, Xia W, Wang HL, Wang YN, Chang WC, Arold ST, Chou CK, Tsou PH, Yamaguchi H, Fang YF, Lee HJ, Lee HH, Tai SK, Yang MH, Morelli MP, Sen M, Ladbury JE, Chen CH, Grandis JR, Kopetz S, Hung MC. PRMT1-mediated methylation of the EGF receptor regulates signaling and cetuximab response. J Clin Invest 2015; 125:4529-43. [PMID: 26571401 DOI: 10.1172/jci82826] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 09/17/2015] [Indexed: 01/08/2023] Open
Abstract
Posttranslational modifications to the intracellular domain of the EGFR are known to regulate EGFR functions; however, modifications to the extracellular domain and their effects remain relatively unexplored. Here, we determined that methylation at R198 and R200 of the EGFR extracellular domain by protein arginine methyltransferase 1 (PRMT1) enhances binding to EGF and subsequent receptor dimerization and signaling activation. In a mouse orthotopic colorectal cancer xenograft model, expression of a methylation-defective EGFR reduced tumor growth. Moreover, increased EGFR methylation sustained signaling activation and cell proliferation in the presence of the therapeutic EGFR monoclonal antibody cetuximab. In colorectal cancer patients, EGFR methylation level also correlated with a higher recurrence rate after cetuximab treatment and reduced overall survival. Together, these data indicate that R198/R200 methylation of the EGFR plays an important role in regulating EGFR functionality and resistance to cetuximab treatment.
Collapse
|
30
|
Etienne-Grimaldi MC, Boyer JC, Thomas F, Quaranta S, Picard N, Loriot MA, Narjoz C, Poncet D, Gagnieu MC, Ged C, Broly F, Le Morvan V, Bouquié R, Gaub MP, Philibert L, Ghiringhelli F, Le Guellec C. UGT1A1genotype and irinotecan therapy: general review and implementation in routine practice. Fundam Clin Pharmacol 2015; 29:219-37. [DOI: 10.1111/fcp.12117] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 02/05/2015] [Accepted: 02/24/2015] [Indexed: 12/19/2022]
Affiliation(s)
| | - Jean-Christophe Boyer
- Unité de Toxicologie; Laboratoire de Biochimie; CHU Carémeau, Place du Pr Debré; 30029 Nîmes Cedex France
| | - Fabienne Thomas
- Institut Claudius Regaud; 1, avenue Irène Joliot-Curie 31059 Toulouse France
| | - Sylvie Quaranta
- Service de Pharmacocinétique et Toxicologie; Laboratoire de Biologie Médicale; Hôpital de la Timone; Bât F; 264 rue Saint Pierre 13385 Marseille Cedex 05 France
| | - Nicolas Picard
- Service Pharmacologie; Toxicologie et Pharmacovigilance; CHU Limoges, Bâtiment CBRS; 2 avenue Martin Luther King 87042 Limoges France
| | - Marie-Anne Loriot
- Hôpital Européen Georges Pompidou; SERVICE BIOCHIMIE; 20 Rue Leblanc 75015 Paris France
| | - Céline Narjoz
- Hôpital Européen Georges Pompidou; SERVICE BIOCHIMIE; 20 Rue Leblanc 75015 Paris France
| | - Delphine Poncet
- Equipe Signalisation Métabolisme et Progression Tumorale; UMR 1052-5286; Centre Léon Bérard; 28 rue Laennec 69373 Lyon Cedex 08 69008 Lyon France
| | - Marie-Claude Gagnieu
- Fédération de Biochimie; UF Pharmacologie Spécialisée; Hôpital E. Herriot; 5 place d'Arsonval 69437 Lyon Cedex 03 France
| | - Cécile Ged
- Plateau Technique de Biologie Moléculaire; Pôle de Biologie et Pathologie; CHU de Bordeaux; 1, place A Raba Leon 33 000 Bordeaux France
| | - Franck Broly
- Service de Toxicologie et Génopathies; Centre de Biologie Pathologie Génétique; Centre Hospitalier Régional et Universitaire de Lille; 59037 Lille Cedex France
| | - Valérie Le Morvan
- Institut Bergonié; Unité Inserm VINCO; 229 cours de l'Argonne 33076 Bordeaux Cedex France
| | - Régis Bouquié
- Laboratoire de Pharmacologie clinique; Institut de Biologie - CHU Nantes; 9, quai Moncousu 44093 Nantes Cedex 1 France
| | - Marie-Pierre Gaub
- EA3430; FMTS Université de Strasbourg; Laboratoire de Biochimie- Biologie Moléculaire; Hôpital de hautepierre; Avenue Molière 67098 Strasbourg France
| | - Laurent Philibert
- Unité de Biopathologie et pharmacogénétique; Laboratoire d'oncopharmacologie; Institut régional du Cancer Montpellier - Val d'Aurelle; 208 Avenue des Apothicaires 34298 Montpellier Cedex 5 France
| | - François Ghiringhelli
- Département de biopathologie; Centre Georges Francois Leclerc; 1 rue du professeur Marion 21000 Dijon France
| | - Chantal Le Guellec
- Unité de pharmacogénétique; Laboratoire de biochimie et biologie moléculaire; CHU Bretonneau; 2 bis boulevard Tonnellé 37044 Tours France
| | | |
Collapse
|
31
|
Hong S, Fang W, Liang W, Yan Y, Zhou T, Qin T, Wu X, Ma Y, Zhao Y, Yang Y, Hu Z, Xue C, Hou X, Chen Y, Huang Y, Zhao H, Zhang L. Risk of treatment-related deaths with vascular endothelial growth factor receptor tyrosine kinase inhibitors: a meta-analysis of 41 randomized controlled trials. Onco Targets Ther 2014; 7:1851-67. [PMID: 25336977 PMCID: PMC4199796 DOI: 10.2147/ott.s68386] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (TKIs) have widely been used in advanced cancer. However, these drugs may also lead to serious adverse events. The present meta-analysis aimed to determine the overall incidence and risk of deaths due to VEGFR-TKIs with more detailed subgroup analysis. Materials and methods PubMed, Web of Science, and Cochrane databases were searched for randomized controlled trials (RCTs) that compared VEGFR-TKIs with non-VEGFR-TKIs in the treatment of solid cancer. Pooled incidence, odds ratios (ORs) and 95% confidence intervals (CIs) were calculated using random-effects or fixed-effects models based on the heterogeneity of included trials. Results A total of 14,139 participants from 41 RCTs were enrolled. The pooled incidence of death due to VEGFR-TKIs was 1.9% (95% CI: 1.6%–2.3%) with an OR of 1.85 (95% CI: 1.33–2.58; P<0.01) when compared with control groups. On subgroup analysis, significantly increased risk of death was found in patients with nonsmall-cell lung cancer (OR: 2.37; 95% CI: 1.19–4.73; P=0.01) and colorectal cancer (OR: 2.84; 95% CI: 1.02–7.96; P=0.05). Among different VEGFR-TKIs, sorafenib and sunitinib had significant risk of death when compared with control arms, respectively. VEGFR-TKIs in combination with other antineoplastic agents, but not VEGFR-TKI monotherapy, significantly increased the risk of treatment-related deaths. No heterogeneity was noted across all the prespecified subgroups regarding ORs. Conclusion The present work pointed out a significantly increased risk of death due to VEGFR-TKIs. Close monitoring should be emphasized in patients receiving these drugs.
Collapse
Affiliation(s)
- Shaodong Hong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
| | - Wenfeng Fang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
| | - Wenhua Liang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
| | - Yue Yan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
| | - Ting Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
| | - Tao Qin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
| | - Xuan Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
| | - Yuxiang Ma
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
| | - Yuanyuan Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
| | - Yunpeng Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
| | - Zhihuang Hu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
| | - Cong Xue
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
| | - Xue Hou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
| | - Yue Chen
- Medical School, University of South China, Hengyang, Hunan, People's Republic of China
| | - Yan Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
| | - Hongyun Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
32
|
Galsky MD, Posner M, Holcombe RF, Lee KM, Misiukiewicz K, Tsao CK, Godbold J, Soto R, Gimpel-Tetra K, Lowe N, Oh WK. Phase Ib study of dovitinib in combination with gemcitabine plus cisplatin or gemcitabine plus carboplatin in patients with advanced solid tumors. Cancer Chemother Pharmacol 2014; 74:465-71. [PMID: 25023489 DOI: 10.1007/s00280-014-2518-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 06/27/2014] [Indexed: 11/29/2022]
Abstract
PURPOSE Dovitinib is a small molecule kinase inhibitor with activity against the fibroblast growth factor and vascular endothelial growth factor receptor families. The purpose of this phase Ib study was to define the recommended phase 2 dose of the combinations of gemcitabine and cisplatin or gemcitabine and carboplatin plus dovitinib. METHODS Patients with advanced solid tumors were enrolled in two parallel dose escalation arms (cisplatin- or carboplatin-based regimens). Treatment was administered with gemcitabine (1,000 mg/m(2) on days 1 and 8), cisplatin (70 mg/m(2)), or carboplatin (AUC 5) on day 1, and dovitinib (orally on days 1-5, 8-12, and 15-19), every 21 days. The starting dose of dovitinib was 300 mg and was dose escalated in successive cohorts using 3 + 3 dose escalation rules. RESULTS Fourteen patients with advanced solid tumors were enrolled, five to the cisplatin arm and nine to the carboplatin arm. Patients enrolled in the cisplatin arm received a median of two cycles of treatment (range 1-5), and patients enrolled in the carboplatin arm received a median of one cycle of treatment (range 1-4). There were no protocol-defined dose-limiting toxicities in the cisplatin arm. However, the cohort was closed due to the need for frequent dose delays and/or reductions and two patients experiencing severe thromboembolic events. There were two dose-limiting toxicities in the carboplatin arm at the starting dose level of dovitinib (both prolonged neutropenia), and the dose of dovitinib was de-escalated to 200 mg. Two additional dose-limiting toxicities (prolonged neutropenia and febrile neutropenia) occurred in the lower dose cohort, and the study was closed. No patients achieved an objective response to treatment. CONCLUSIONS Dovitinib in combination with gemcitabine plus cisplatin or gemcitabine plus carboplatin was poorly tolerated due to myelosuppression.
Collapse
Affiliation(s)
- Matthew D Galsky
- Division of Hematology/Oncology, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|