1
|
Martin-Broto J, Diaz-Beveridge R, Moura D, Ramos R, Martinez-Trufero J, Carrasco I, Sebio A, González-Billalabeitia E, Gutierrez A, Fernandez-Jara J, Hernández-Vargas L, Cruz J, Valverde C, Hindi N. Phase Ib Study for the Combination of Doxorubicin, Dacarbazine, and Nivolumab as the Upfront Treatment in Patients With Advanced Leiomyosarcoma: A Study by the Spanish Sarcoma Group (GEIS). J Clin Oncol 2025; 43:297-307. [PMID: 39356980 DOI: 10.1200/jco.24.00358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/06/2024] [Accepted: 07/17/2024] [Indexed: 10/04/2024] Open
Abstract
PURPOSE Doxorubicin, alongside a select group of cytotoxic agents, is capable of inducing an adaptive immune response via a well-established peculiar type of tumor cell death called immunogenic cell death (ICD). We hypothesize that combining doxorubicin and dacarbazine with nivolumab may enhance therapeutic efficacy by exerting synergy in the ICD circuit. We hereby present a phase Ib trial with this combination. PATIENTS AND METHODS Patients with advanced leiomyosarcoma and anthracycline-naïve were eligible. The initial dose level consisted of doxorubicin 75 mg/m2 once on day 1, once every three weeks, followed by dacarbazine 400 mg/m2 once on days 1 and 2, once every three weeks, plus nivolumab 360 mg once on day 2, once every 3 weeks, for six courses and then 1 year of nivolumab. A (-1) dose level was the same regimen but with nivolumab 240 mg. A classic 3 + 3 phase-I design was used to determine the recommended phase-II dose (RP2D). Secondary end points included overall response rate, safety profile, survival, and translational research. RESULTS From January 2002 to July 2023, 24 patients were enrolled and 23 were evaluable for efficacy, excluding one patient because of noncompliant dose. All patients were treated with the initial dose level, then the RP2D. Toxicity was mild, with the most frequent being grade 4 toxicity neutropenia (16.7%) and thrombocytopenia (8.3%), while no grade 5 toxicity occurred. The centrally reviewed objective response rate was as follows: partial response 56.5%, stable disease 39.1%, and progression 4.4%. The 6-month progression-free survival (PFS) rate was 80% (95% CI, 63 to 98). Dynamic increases of HMGB1 in blood significantly correlated with longer PFS. CONCLUSION This scheme of doxorubicin, dacarbazine, and nivolumab is feasible and well tolerated. Clinical activity is encouraging and the prognostic impact of HMGB1 supports the relevance of ICD activation. Further clinical research is already underway with this concept in leiomyosarcoma.
Collapse
Affiliation(s)
- Javier Martin-Broto
- Medical Oncology Department, Fundacion Jimenez Diaz University Hospital, University Hospital General de Villalba, Madrid, Spain
- Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| | | | - David Moura
- Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| | - Rafael Ramos
- Pathology Department, Hospital Universitari Son Espases, Palma de Mallorca, Spain
| | | | - Irene Carrasco
- Medical Oncology Department, Hospital Universitario Virgen del Rocio, Sevilla, Spain
| | - Ana Sebio
- Medical Oncology Department, Hospital Sant Pau, Barcelona, Spain
| | | | - Antonio Gutierrez
- Hematology Department, Hospital Universitari Son Espases, Palma de Mallorca, Spain
| | | | | | - Josefina Cruz
- Medical Oncology Department, Hospital Universitario de Canarias, La Laguna, Spain
| | - Claudia Valverde
- Medical Oncology Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Nadia Hindi
- Medical Oncology Department, Fundacion Jimenez Diaz University Hospital, University Hospital General de Villalba, Madrid, Spain
- Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| |
Collapse
|
2
|
Roets E, van der Graaf W, van Riet BHG, Haas RL, Younger E, Sparano F, Wilson R, van der Mierden S, Steeghs N, Efficace F, Husson O. Patient-reported outcomes in randomized clinical trials of systemic therapy for advanced soft tissue sarcomas in adults: A systematic review. Crit Rev Oncol Hematol 2024; 197:104345. [PMID: 38582227 DOI: 10.1016/j.critrevonc.2024.104345] [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: 01/05/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND This systematic review evaluates reporting of patient-reported outcomes (PROs) within randomized clinical trials (RCTs) for advanced soft tissue sarcoma (STS) patients. METHODS A systematic literature search from January 2000 - August 2022 was conducted for phase II/III RCTs evaluating systemic treatments in adult patients with advanced STS. Quality of PRO reporting was assessed using the CONSORT PRO extension. RESULTS Out of 7294 abstracts, 59 articles were included; comprising 43 RCTs. Only 15 RCTs (35%) included PROs, none as primary endpoints. Only 10 of these RCTs reported PROs, either in the primary (6/10) or secondary publication (1/10) or in both (3/10), with a median time interval of 23 months. The median CONSORT PRO adherence score was 5.5/14, with higher scores in publications focusing exclusively on PROs. CONCLUSION These results highlight the need for improved and more consistent PRO reporting to inform patient care in the setting of advanced STS.
Collapse
Affiliation(s)
- Evelyne Roets
- Department of Medical Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, the Netherlands
| | - Winette van der Graaf
- Department of Medical Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, the Netherlands; Department of Medical Oncology, ErasmusMC Cancer Institute, Erasmus University Medical Center, Doctor Molewaterplein 40, Rotterdam 3015 GD, the Netherlands
| | - Bauke H G van Riet
- Department of Radiotherapy, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, the Netherlands
| | - Rick L Haas
- Department of Radiotherapy, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, the Netherlands; Department of Radiotherapy, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, the Netherlands
| | - Eugenie Younger
- Sarcoma Unit, Royal Marsden NHS Foundation Trust, Fulham Road, London SW3 6JJ, United Kingdom
| | - Francesco Sparano
- Health Outcomes Research Unit, Italian Group for Adult Hematologic Diseases (GIMEMA) Data Center, Rome, Italy
| | - Roger Wilson
- Sarcoma Patients Advocacy Global Network, Untergasse 36, Wölfersheim D-61200, Germany; Sarcoma UK, 17/18 Angel Gate, City Road, London, UK
| | - Stevie van der Mierden
- Scientific information service, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, the Netherlands
| | - Neeltje Steeghs
- Department of Medical Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, the Netherlands
| | - Fabio Efficace
- Health Outcomes Research Unit, Italian Group for Adult Hematologic Diseases (GIMEMA) Data Center, Rome, Italy
| | - Olga Husson
- Department of Medical Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, the Netherlands; Department of Surgical Oncology, ErasmusMC Cancer Institute, Erasmus University Medical Center, Doctor Molewaterplein 40, Rotterdam 3015 GD, the Netherlands.
| |
Collapse
|
3
|
Tian Z, Yao W. Chemotherapeutic drugs for soft tissue sarcomas: a review. Front Pharmacol 2023; 14:1199292. [PMID: 37637411 PMCID: PMC10450752 DOI: 10.3389/fphar.2023.1199292] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 08/03/2023] [Indexed: 08/29/2023] Open
Abstract
Despite the low incidence of soft tissue sarcomas (STSs), hundreds of thousands of new STS cases are diagnosed annually worldwide, and approximately half of them eventually progress to advanced stages. Currently, chemotherapy is the first-line treatment for advanced STSs. There are difficulties in selecting appropriate drugs for multiline chemotherapy, or for combination treatment of different STS histological subtypes. In this study, we first comprehensively reviewed the efficacy of various chemotherapeutic drugs in the treatment of STSs, and then described the current status of sensitive drugs for different STS subtypes. anthracyclines are the most important systemic treatment for advanced STSs. Ifosfamide, trabectedin, gemcitabine, taxanes, dacarbazine, and eribulin exhibit certain activities in STSs. Vinca alkaloid agents (vindesine, vinblastine, vinorelbine, vincristine) have important therapeutic effects in specific STS subtypes, such as rhabdomyosarcoma and Ewing sarcoma family tumors, whereas their activity in other subtypes is weak. Other chemotherapeutic drugs (methotrexate, cisplatin, etoposide, pemetrexed) have weak efficacy in STSs and are rarely used. It is necessary to select specific second- or above-line chemotherapeutic drugs depending on the histological subtype. This review aims to provide a reference for the selection of chemotherapeutic drugs for multi-line therapy for patients with advanced STSs who have an increasingly long survival.
Collapse
Affiliation(s)
| | - Weitao Yao
- Department of Orthopedics, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
4
|
Teo AYT, Lim VY, Yang VS. MicroRNAs in the Pathogenesis, Prognostication and Prediction of Treatment Resistance in Soft Tissue Sarcomas. Cancers (Basel) 2023; 15:cancers15030577. [PMID: 36765536 PMCID: PMC9913386 DOI: 10.3390/cancers15030577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/19/2023] Open
Abstract
Soft tissue sarcomas are highly aggressive malignant neoplasms of mesenchymal origin, accounting for less than 1% of adult cancers, but comprising over 20% of paediatric solid tumours. In locally advanced, unresectable, or metastatic disease, outcomes from even the first line of systemic treatment are invariably poor. MicroRNAs (miRNAs), which are short non-coding RNA molecules, target and modulate multiple dysregulated target genes and/or signalling pathways within cancer cells. Accordingly, miRNAs demonstrate great promise for their utility in diagnosing, prognosticating and improving treatment for soft tissue sarcomas. This review aims to provide an updated discussion on the known roles of specific miRNAs in the pathogenesis of sarcomas, and their potential use in prognosticating outcomes and prediction of therapeutic resistance.
Collapse
Affiliation(s)
- Andrea York Tiang Teo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Vivian Yujing Lim
- Institute of Molecular and Cell Biology, A*STAR, Singapore 138673, Singapore
| | - Valerie Shiwen Yang
- Institute of Molecular and Cell Biology, A*STAR, Singapore 138673, Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore
- Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Correspondence:
| |
Collapse
|
5
|
Vasella M, Gousopoulos E, Guidi M, Storti G, Song SY, Grieb G, Pauli C, Lindenblatt N, Giovanoli P, Kim BS. Targeted therapies and checkpoint inhibitors in sarcoma. QJM 2022; 115:793-805. [PMID: 33486519 DOI: 10.1093/qjmed/hcab014] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 01/11/2021] [Indexed: 12/15/2022] Open
Abstract
Sarcomas are defined as a group of mesenchymal malignancies with over 100 heterogeneous subtypes. As a rare and difficult to diagnose entity, micrometastasis is already present at the time of diagnosis in many cases. Current treatment practice of sarcomas consists mainly of surgery, (neo)adjuvant chemo- and/or radiotherapy. Although the past decade has shown that particular genetic abnormalities can promote the development of sarcomas, such as translocations, gain-of-function mutations, amplifications or tumor suppressor gene losses, these insights have not led to established alternative treatment strategies so far. Novel therapeutic concepts with immunotherapy at its forefront have experienced some remarkable success in different solid tumors while their impact in sarcoma remains limited. In this review, the most common immunotherapy strategies in sarcomas, such as immune checkpoint inhibitors, targeted therapy and cytokine therapy are concisely discussed. The programmed cell death (PD)-1/PD-1L axis and apoptosis-inducing cytokines, such as TNF-related apoptosis-inducing ligand (TRAIL), have not yielded the same success like in other solid tumors. However, in certain sarcoma subtypes, e.g. liposarcoma or undifferentiated pleomorphic sarcoma, encouraging results in some cases when employing immune checkpoint inhibitors in combination with other treatment options were found. Moreover, newer strategies such as the targeted therapy against the ancient cytokine macrophage migration inhibitory factor (MIF) may represent an interesting approach worth investigation in the future.
Collapse
Affiliation(s)
- M Vasella
- From the Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - E Gousopoulos
- From the Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - M Guidi
- From the Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - G Storti
- Department of Surgical Sciences, Plastic and Reconstructive Surgery, University of Rome-'Tor Vergata', Via Montepellier, 1, 00133 Rome, Italy
| | - S Y Song
- Department of Plastic and Reconstructive Surgery, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, Korea
| | - G Grieb
- Department of Plastic Surgery and Hand Surgery, Gemeinschaftskrankenhaus Havelhoehe, Kladower Damm 221, 14089 Berlin, Germany
- Department of Plastic Surgery, Hand Surgery and Burn Center, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - C Pauli
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - N Lindenblatt
- From the Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - P Giovanoli
- From the Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - B-S Kim
- From the Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| |
Collapse
|
6
|
Targeting TRAIL Death Receptors in Triple-Negative Breast Cancers: Challenges and Strategies for Cancer Therapy. Cells 2022; 11:cells11233717. [PMID: 36496977 PMCID: PMC9739296 DOI: 10.3390/cells11233717] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/11/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
The tumor necrosis factor (TNF) superfamily member TNF-related apoptosis-inducing ligand (TRAIL) induces apoptosis in cancer cells via death receptor (DR) activation with little toxicity to normal cells or tissues. The selectivity for activating apoptosis in cancer cells confers an ideal therapeutic characteristic to TRAIL, which has led to the development and clinical testing of many DR agonists. However, TRAIL/DR targeting therapies have been widely ineffective in clinical trials of various malignancies for reasons that remain poorly understood. Triple negative breast cancer (TNBC) has the worst prognosis among breast cancers. Targeting the TRAIL DR pathway has shown notable efficacy in a subset of TNBC in preclinical models but again has not shown appreciable activity in clinical trials. In this review, we will discuss the signaling components and mechanisms governing TRAIL pathway activation and clinical trial findings discussed with a focus on TNBC. Challenges and potential solutions for using DR agonists in the clinic are also discussed, including consideration of the pharmacokinetic and pharmacodynamic properties of DR agonists, patient selection by predictive biomarkers, and potential combination therapies. Moreover, recent findings on the impact of TRAIL treatment on the immune response, as well as novel strategies to address those challenges, are discussed.
Collapse
|
7
|
WNT/β-Catenin Pathway in Soft Tissue Sarcomas: New Therapeutic Opportunities? Cancers (Basel) 2021; 13:cancers13215521. [PMID: 34771683 PMCID: PMC8583315 DOI: 10.3390/cancers13215521] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The WNT/β-catenin signaling pathway is involved in fundamental processes for the proliferation and differentiation of mesenchymal stem cells. However, little is known about its relevance for mesenchymal neoplasms, such us soft tissue sarcomas (STS). Chemotherapy based on doxorubicin (DXR) still remains the standard first-line treatment for locally advanced unresectable or metastatic STS, although overall survival could not be improved by combination with other chemotherapeutics. In this sense, the development of new therapeutic approaches continues to be an unmatched goal. This review covers the most important molecular alterations of the WNT signaling pathway in STS, broadening the current knowledge about STS as well as identifying novel drug targets. Furthermore, the current therapeutic options and drug candidates to modulate WNT signaling, which are usually classified by their interaction site upstream or downstream of β-catenin, and their presumable clinical impact on STS are discussed. Abstract Soft tissue sarcomas (STS) are a very heterogeneous group of rare tumors, comprising more than 50 different histological subtypes that originate from mesenchymal tissue. Despite their heterogeneity, chemotherapy based on doxorubicin (DXR) has been in use for forty years now and remains the standard first-line treatment for locally advanced unresectable or metastatic STS, although overall survival could not be improved by combination with other chemotherapeutics. In this sense, the development of new therapeutic approaches continues to be a largely unmatched goal. The WNT/β-catenin signaling pathway is involved in various fundamental processes for embryogenic development, including the proliferation and differentiation of mesenchymal stem cells. Although the role of this pathway has been widely researched in neoplasms of epithelial origin, little is known about its relevance for mesenchymal neoplasms. This review covers the most important molecular alterations of the WNT signaling pathway in STS. The detection of these alterations and the understanding of their functional consequences for those pathways controlling sarcomagenesis development and progression are crucial to broaden the current knowledge about STS as well as to identify novel drug targets. In this regard, the current therapeutic options and drug candidates to modulate WNT signaling, which are usually classified by their interaction site upstream or downstream of β-catenin, and their presumable clinical impact on STS are also discussed.
Collapse
|
8
|
Hua Q, Xu G, Zhao L, Zhang T. Comparison of first line chemotherapy regimens for advanced soft tissue sarcoma: a network meta-analysis. J Chemother 2021; 33:570-581. [PMID: 33870875 DOI: 10.1080/1120009x.2021.1913703] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The best first line chemotherapy regimen for advanced soft tissue sarcoma (ASTS) remains inconclusive. Here, we aimed to find the best first line chemotherapy regimen by performing a network meta-analysis. Regimens were compared in terms of overall survival (OS), overall response rate(ORR), progression free survival (PFS), and toxicity. Twenty-eight eligible trials with a total of 6928 patients were included. EC (epirubicin + cisplatin) was considered as the better regimen for advanced STS with probability of 61.9% in terms of OS. However, this regimen only have been evaluated in a single small trial and tend to have more hematological toxicities than doxorubicin. No regimen was superior to doxorubicin with significant statistical difference in terms of PFS and ORR, even aldoxorubicin behaved better than doxorubicin in the network analysis. Collectively, doxorubicin still can be selected preferentially for the first line chemotherapy for patients.
Collapse
Affiliation(s)
- Qingling Hua
- Cancer center, Union hospital, Tongji medical college, Huazhong university of science and technology, Wuhan, China
| | - Guojie Xu
- Cancer center, Union hospital, Tongji medical college, Huazhong university of science and technology, Wuhan, China
| | - Lei Zhao
- Cancer center, Union hospital, Tongji medical college, Huazhong university of science and technology, Wuhan, China
| | - Tao Zhang
- Cancer center, Union hospital, Tongji medical college, Huazhong university of science and technology, Wuhan, China
| |
Collapse
|
9
|
Apoptosis-Inducing TNF Superfamily Ligands for Cancer Therapy. Cancers (Basel) 2021; 13:cancers13071543. [PMID: 33801589 PMCID: PMC8036978 DOI: 10.3390/cancers13071543] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/21/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer is a complex disease with apoptosis evasion as one of its hallmarks; therefore, apoptosis induction in transformed cells seems a promising approach as a cancer treatment. TNF apoptosis-inducing ligands, which are naturally present in the body and possess tumoricidal activity, are attractive candidates. The most studied proteins are TNF-α, FasL, and TNF-related apoptosis-inducing ligand (TRAIL). Over the years, different recombinant TNF family-derived apoptosis-inducing ligands and agonists have been designed. Their stability, specificity, and half-life have been improved because most of the TNF ligands have the disadvantages of having a short half-life and affinity to more than one receptor. Here, we review the outlook on apoptosis-inducing ligands as cancer treatments in diverse preclinical and clinical stages and summarize strategies of overcoming their natural limitations to improve their effectiveness.
Collapse
|
10
|
Bebb DG, Banerji S, Blais N, Desmeules P, Gill S, Grin A, Feilotter H, Hansen AR, Hyrcza M, Krzyzanowska M, Melosky B, Noujaim J, Purgina B, Ruether D, Simmons CE, Soulieres D, Torlakovic EE, Tsao MS. Canadian Consensus for Biomarker Testing and Treatment of TRK Fusion Cancer in Adults. Curr Oncol 2021; 28:523-548. [PMID: 33467570 PMCID: PMC7903287 DOI: 10.3390/curroncol28010053] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/31/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
The tyrosine receptor kinase (TRK) inhibitors larotrectinib and entrectinib were recently approved in Canada for the treatment of solid tumours harbouring neurotrophic tyrosine receptor kinase (NTRK) gene fusions. These NTRK gene fusions are oncogenic drivers found in most tumour types at a low frequency (<5%), and at a higher frequency (>80%) in a small number of rare tumours (e.g., secretory carcinoma of the salivary gland and of the breast). They are generally mutually exclusive of other common oncogenic drivers. Larotrectinib and entrectinib have demonstrated impressive overall response rates and tolerability in Phase I/II trials in patients with TRK fusion cancer with no other effective treatment options. Given the low frequency of TRK fusion cancer and the heterogeneous molecular testing landscape in Canada, identifying and optimally managing such patients represents a new challenge. We provide a Canadian consensus on when and how to test for NTRK gene fusions and when to consider treatment with a TRK inhibitor. We focus on five tumour types: thyroid carcinoma, colorectal carcinoma, non-small cell lung carcinoma, soft tissue sarcoma, and salivary gland carcinoma. Based on the probability of the tumour harbouring an NTRK gene fusion, we also suggest a tumour-agnostic consensus for NTRK gene fusion testing and treatment. We recommend considering a TRK inhibitor in all patients with TRK fusion cancer with no other effective treatment options.
Collapse
Affiliation(s)
- D. Gwyn Bebb
- Tom Baker Cancer Centre and University of Calgary, Calgary, AB T2N 4N2, Canada
| | - Shantanu Banerji
- Research Institute in Oncology and Hematology, CancerCare Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada;
| | - Normand Blais
- Centre Hospitalier Universitaire de Montreal, Department of Medicine, University of Montreal, Montreal, QC H2X 3E4, Canada; (N.B.); (D.S.)
| | - Patrice Desmeules
- Service D’Anatomopathologie et de Cytologie, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, QC G1V 0A6, Canada;
| | - Sharlene Gill
- BC Cancer, Vancouver, BC V5Z 4E6, Canada; (S.G.); (B.M.); (C.E.S.)
| | - Andrea Grin
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada; (A.G.); (H.F.)
| | - Harriet Feilotter
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada; (A.G.); (H.F.)
| | - Aaron R. Hansen
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; (A.R.H.); (M.K.)
| | - Martin Hyrcza
- Department of Pathology and Laboratory Medicine, Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB T2N 4Z6, Canada;
| | - Monika Krzyzanowska
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; (A.R.H.); (M.K.)
| | - Barbara Melosky
- BC Cancer, Vancouver, BC V5Z 4E6, Canada; (S.G.); (B.M.); (C.E.S.)
| | | | - Bibiana Purgina
- The Ottawa Hospital, Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, ON K1N 6N5, Canada;
| | - Dean Ruether
- Department of Oncology, Tom Baker Cancer Centre, Calgary, AB T2N 4N2, Canada;
| | | | - Denis Soulieres
- Centre Hospitalier Universitaire de Montreal, Department of Medicine, University of Montreal, Montreal, QC H2X 3E4, Canada; (N.B.); (D.S.)
| | - Emina Emilia Torlakovic
- Department of Pathology and Laboratory Medicine, Saskatchewan Health Authority and University of Saskatchewan, Saskatoon, SK S7N 5B5, Canada;
| | - Ming-Sound Tsao
- Department of Pathology, Laboratory Medicine Program, University Health Network, Toronto, ON M5G 2C4, Canada
| |
Collapse
|
11
|
Martin-Broto J, Hindi N, Lopez-Pousa A, Peinado-Serrano J, Alvarez R, Alvarez-Gonzalez A, Italiano A, Sargos P, Cruz-Jurado J, Isern-Verdum J, Dolado MC, Rincon-Pérez I, Sanchez-Bustos P, Gutierrez A, Romagosa C, Morosi C, Grignani G, Gatti M, Luna P, Alastuey I, Redondo A, Belinchon B, Martinez-Serra J, Sunyach MP, Coindre JM, Dei Tos AP, Romero J, Gronchi A, Blay JY, Moura DS. Assessment of Safety and Efficacy of Combined Trabectedin and Low-Dose Radiotherapy for Patients With Metastatic Soft-Tissue Sarcomas: A Nonrandomized Phase 1/2 Clinical Trial. JAMA Oncol 2020; 6:535-541. [PMID: 32077895 DOI: 10.1001/jamaoncol.2019.6584] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Importance Active therapeutic combinations, such as trabectedin and radiotherapy, offer potentially higher dimensional response in second-line treatment of advanced soft-tissue sarcomas. Dimensional response can be relevant both for symptom relief and for survival. Objective To assess the combined use of trabectedin and radiotherapy in treating patients with progressing metastatic soft-tissue sarcomas. Design, Setting, and Participants Phase 1 of this nonrandomized clinical trial followed the classic 3 + 3 design, with planned radiotherapy at a fixed dose of 30 Gy (3 Gy/d for 10 days) and infusion of trabectedin at 1.3 mg/m2 as the starting dose, 1.5 mg/m2 as dose level +1, and 1.1 mg/m2 as dose level -1. Phase 2 followed the Simon optimal 2-stage design. Allowing for type I and II errors of 10%, treatment success was defined as an overall response rate of 35%. This study was conducted in 9 sarcoma referral centers in Spain, France, and Italy from April 13, 2015, to November 20, 2018. Adult patients with progressing metastatic soft-tissue sarcoma and having undergone at least 1 previous line of systemic therapy were enrolled. In phase 2, patients fitting inclusion criteria and receiving at least 1 cycle of trabectedin and the radiotherapy regimen constituted the per-protocol population; those receiving at least 1 cycle of trabectedin, the safety population. Interventions Trabectedin was administered every 3 weeks in a 24-hour infusion. Radiotherapy was required to start within 1 hour after completion of the first trabectedin infusion (cycle 1, day 2). Main Outcomes and Measures The dose-limiting toxic effects of trabectedin (phase 1) and the overall response rate (phase 2) with use of trabectedin plus irradiation in metastatic soft-tissue sarcomas. Results Eighteen patients (11 of whom were male) were enrolled in phase 1, and 27 other patients (14 of whom were female) were enrolled in phase 2. The median ages of those enrolled in phases 1 and 2 were 42 (range, 23-74) years and 51 (range, 27-73) years, respectively. In phase 1, dose-limiting toxic effects included grade 4 neutropenia lasting more than 5 days in 1 patient at the starting dose level and a grade 4 alanine aminotransferase level increase in 1 of 6 patients at the +1 dose level. In phase 2, among 25 patients with evaluable data, the overall response rate was 72% (95% CI, 53%-91%) for local assessment and 60% (95% CI, 39%-81%) for central assessment. Conclusions and Relevance The findings of this study suggest that the recommended dose of trabectedin for use in combination with this irradiation regimen is 1.5 mg/m2. The trial met its primary end point, with a high overall response rate that indicates the potential of this combination therapy for achieving substantial tumor shrinkage beyond first-line systemic therapy in patients with metastatic, progressing soft-tissue sarcomas. Trial Registration ClinicalTrials.gov Identifier: NCT02275286.
Collapse
Affiliation(s)
- Javier Martin-Broto
- Department of Medical Oncology, University Hospital Virgen del Rocío, Sevilla, Spain.,TERABIS Group, IBiS (Instituto de Biomedicina de Sevilla), Sevilla, Spain
| | - Nadia Hindi
- Department of Medical Oncology, University Hospital Virgen del Rocío, Sevilla, Spain.,TERABIS Group, IBiS (Instituto de Biomedicina de Sevilla), Sevilla, Spain
| | - Antonio Lopez-Pousa
- Department of Medical Oncology, Santa Creu i Sant Pau Hospital, Barcelona, Spain
| | - Javier Peinado-Serrano
- TERABIS Group, IBiS (Instituto de Biomedicina de Sevilla), Sevilla, Spain.,CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Instituto de Salud Carlos III, Madrid, Spain.,Department of Radiation Oncology, University Hospital Virgen del Rocío, Sevilla, Spain
| | - Rosa Alvarez
- Department of Medical Oncology, Gregorio Marañon University Hospital, Madrid, Spain
| | | | - Antoine Italiano
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - Paul Sargos
- Department of Radiotherapy, Institut Bergonié, Bordeaux, France
| | - Josefina Cruz-Jurado
- Department of Medical Oncology, University Hospital of the Canary Islands, Tenerife, Spain
| | | | - Maria Carmen Dolado
- Department of Radiation Oncology, University Hospital of the Canary Islands, Tenerife, Spain
| | | | | | - Antonio Gutierrez
- Department of Hematology, University Hospital Son Espases, Mallorca, Spain
| | - Cleofe Romagosa
- Department of Pathology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Carlo Morosi
- Department of Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giovanni Grignani
- Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Marco Gatti
- Division of Radiotherapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Pablo Luna
- Department of Medical Oncology, University Hospital Son Espases, Mallorca, Spain
| | - Ignacio Alastuey
- Radiotherapy Department, University Hospital Son Espases, Mallorca, Spain
| | - Andres Redondo
- Medical Oncology Department, University Hospital La Paz, Madrid, Spain.,Health Research Institute of La Paz Hospital (IdiPAZ), Madrid, Spain
| | - Belen Belinchon
- Department of Radiotherapy, University Hospital La Paz, Madrid, Spain
| | | | | | - Jean-Michel Coindre
- Department of Biopathology, Institut Bergonié, Bordeaux, France.,Department of Biopathology, Bordeaux University, Talence, France
| | - Angelo P Dei Tos
- Department of Medicine, University of Padua School of Medicine, Padua, Italy
| | - Jesus Romero
- Department of Radiation Oncology, University Hospital Puerta de Hierro, Madrid, Spain
| | - Alessandro Gronchi
- Department of Surgery, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Jean-Yves Blay
- Medical Oncology Department, Centre Léon Bérard, Lyon, France.,Département of Medicine, Université Claude Bernard Lyon I, Lyon, France
| | - David S Moura
- TERABIS Group, IBiS (Instituto de Biomedicina de Sevilla), Sevilla, Spain
| |
Collapse
|
12
|
Smolle MA, Szkandera J, Andreou D, Palmerini E, Bergovec M, Leithner A. Treatment options in unresectable soft tissue and bone sarcoma of the extremities and pelvis - a systematic literature review. EFORT Open Rev 2020; 5:799-814. [PMID: 33312707 PMCID: PMC7722943 DOI: 10.1302/2058-5241.5.200069] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
In patients with metastatic or unresectable soft tissue and bone sarcoma of extremities and pelvis, survival is generally poor. The aim of the current systematic review was to analyse recent publications on treatment approaches in patients with inoperable and/or metastatic sarcoma. Original articles published between 1st January 2011 and 2nd May 2020, using the search terms ‘unresectable sarcoma’, ‘inoperability AND sarcoma’, ‘inoperab* AND sarcoma’, and ‘treatment AND unresectable AND sarcoma’ in PubMed, were potentially eligible. Out of the 839 initial articles (containing 274 duplicates) obtained and 23 further articles identified by cross-reference checking, 588 were screened, of which 447 articles were removed not meeting the inclusion criteria. A further 54 articles were excluded following full-text assessment, resulting in 87 articles finally being analysed. Of the 87 articles, 38 were retrospective (43.7%), two prospective (2.3%), six phase I or I/II trials (6.9%), 22 phase II non-randomized trials (27.6%), nine phase II randomized trials (10.3%) and eight phase III randomized trials (9.2%). Besides radio/particle therapy, isolated limb perfusion and conventional chemotherapy, novel therapeutic approaches, including immune checkpoint inhibitors and tyrosine kinase inhibitors were also identified, with partially very promising effects in advanced sarcomas. Management of inoperable, advanced or metastatic sarcomas of the pelvis and extremities remains challenging, with the optimal treatment to be defined individually. Besides conventional chemotherapy, some novel therapeutic approaches have promising effects in both bone and soft tissue subtypes. Considering that only a small proportion of studies were randomized, the clinical evidence currently remains moderate and thus calls for further large, randomized clinical trials.
Cite this article: EFORT Open Rev 2020;5:799-814. DOI: 10.1302/2058-5241.5.200069
Collapse
Affiliation(s)
- Maria Anna Smolle
- Department of Orthopaedics and Trauma, Medical University of Graz, Graz, Austria
| | - Joanna Szkandera
- Division of Clinical Oncology, Internal Medicine, Medical University of Graz, Graz, Austria
| | - Dimosthenis Andreou
- Division of Orthopaedic Oncology and Sarcoma Surgery, Helios Klinikum Bad Saarow, Sarcoma Center Berlin-Brandenburg, Berlin, Germany
| | - Emanuela Palmerini
- Chemotherapy Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna University, Bologna, Italy
| | - Marko Bergovec
- Department of Orthopaedics and Trauma, Medical University of Graz, Graz, Austria
| | - Andreas Leithner
- Department of Orthopaedics and Trauma, Medical University of Graz, Graz, Austria
| |
Collapse
|
13
|
Yamamoto J, Miyake K, Han Q, Tan Y, Inubushi S, Sugisawa N, Higuchi T, Tashiro Y, Nishino H, Homma Y, Matsuyama R, Chawla SP, Bouvet M, Singh SR, Endo I, Hoffman RM. Oral recombinant methioninase increases TRAIL receptor-2 expression to regress pancreatic cancer in combination with agonist tigatuzumab in an orthotopic mouse model. Cancer Lett 2020; 492:174-184. [PMID: 32739322 DOI: 10.1016/j.canlet.2020.07.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/08/2020] [Accepted: 07/27/2020] [Indexed: 12/16/2022]
Abstract
Methionine addiction is a fundamental and general hallmark of cancer. Gene expression analysis showed that methionine restriction (MR) of methionine-addicted cancer cells increases TNF-related apoptosis-induced ligand receptor-2 (TRAIL-R2) expression. Here, we determined the effects of MR on TRAIL-R2 targeted therapy in pancreatic cancer by the TRAIL-R2 agonist tigatuzumab. Human pancreatic cancer cell lines were cultured in control or methionine-free medium. The effects of MR on TRAIL-R2 expression and sensitivity to tigatuzumab were evaluated in vitro. An orthotopic pancreatic cancer mouse model was established to evaluate the efficacy of MR using oral recombinant methioninase (o-rMETase), and the efficacy of tigatuzumab and their combination. MR enabled tigatuzumab-induced apoptosis, by increasing TRAIL-R2 expression in pancreatic cancer cells in vitro. The protein expression level of the melanoma-associated antigen MAGED2, which reduces TRAIL-R2 expression, was decreased by MR. In the orthotopic pancreatic cancer mouse model, o-rMETase increased TRAIL-R2 expression level in the tumors and enabled the antitumor efficacy of tigatuzumab. MR, effected by o-rMETase, enabled the efficacy of the TRAIL-R2 agonist tigatuzumab by increasing TRAIL-R2 expression in pancreatic cancer. Our results suggest that o-rMETase has clinical potential for treating pancreatic cancer.
Collapse
Affiliation(s)
- Jun Yamamoto
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA; Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kentaro Miyake
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA; Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | | | | | - Sachiko Inubushi
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Norihiko Sugisawa
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Takashi Higuchi
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Yoshihiko Tashiro
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Hiroto Nishino
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Yuki Homma
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ryusei Matsuyama
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | | | - Michael Bouvet
- Department of Surgery, University of California, San Diego, CA, USA
| | - Shree Ram Singh
- Basic Research Laboratory, National Cancer Institute, Frederick, MD, USA.
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Robert M Hoffman
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA.
| |
Collapse
|
14
|
Skelton WP, Turba E, Sokol L. Durable Complete Response to AMG 655 (Conatumumab) and Vorinostat in a Patient With Relapsed Classical Hodgkin Lymphoma: Extraordinary Response from a Phase 1b Clinical Protocol. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e944-e946. [PMID: 32828719 DOI: 10.1016/j.clml.2020.07.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 01/13/2023]
Affiliation(s)
- William Paul Skelton
- Division of Medical Oncology, H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL.
| | - Elyce Turba
- Division of Medical Oncology, H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL
| | - Lubomir Sokol
- Division of Medical Oncology, H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL.
| |
Collapse
|
15
|
Lee EY, Yu JY, Paek AR, Lee SH, Jang H, Cho SY, Kim JH, Kang HG, Yun T, Oh SE, Park SY, You HJ. Targeting TJP1 attenuates cell-cell aggregation and modulates chemosensitivity against doxorubicin in leiomyosarcoma. J Mol Med (Berl) 2020; 98:761-773. [PMID: 32318747 DOI: 10.1007/s00109-020-01909-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/30/2020] [Accepted: 04/03/2020] [Indexed: 12/24/2022]
Abstract
Tight junction protein 1 (TJP1) is a membrane-associated cytosolic protein important for cell-cell communication in intercellular barriers in epithelial and non-epithelial cells. Here, we explored the functional involvement of TJP1 in non-epithelial tumors such as soft tissue sarcoma, especially in leiomyosarcoma (LMS). TJP1 expression in soft tissue sarcoma was analyzed in normal and tumor tissues as well as from public datasets such as the TCGA provisional dataset, in which TJP1 expression was compared with other subtypes such as undifferentiated sarcomas, and myxofibrosarcomas. SK-LMS-1 cell lines with reduced TJP1 expression showed attenuated anchorage-independent colony formation as well as reduced intercellular aggregation on non-coated culture plates compared with control as well as parental SK-LMS-1 cells. Transcriptome profiling following TJP1 knockdown in SK-LMS-1 cells suggested the involvement of several signaling pathways, including NF-κB pathway and growth factor receptor signaling. In addition, TJP1 downregulation induced enhanced response against anti-cancer agents, doxorubicin and gefitinib. Taken together, these results suggest that TJP1 contributes to sarcoma genesis and might be useful therapeutic target. KEY MESSAGES: • TJP1 expression at RNA level higher in tumor than in normal tissues of sarcoma. • Targeting TJP1 attenuates cell-cell aggregation and anchorage-independent growth. • Targeting TJP1 is beneficial in anti-cancer therapy in LMS.
Collapse
Affiliation(s)
- Eun-Young Lee
- Division of Translational Science, Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang, Gyeonggi, 10408, South Korea.,Department of Medical Biotechnology, Yeungnam University, 280 Daehak-ro, Gyeongsan, 38541, South Korea
| | - Jung Yeon Yu
- Division of Translational Science, Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang, Gyeonggi, 10408, South Korea
| | - A Rome Paek
- Division of Translational Science, Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang, Gyeonggi, 10408, South Korea
| | - So Hee Lee
- Division of Translational Science, Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang, Gyeonggi, 10408, South Korea
| | - Hyonchol Jang
- Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy (NCC-GCSP), National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang, Gyeonggi, 10408, South Korea.,Division of Cancer Biology, Research Institute, National Cancer Center, Goyang, South Korea
| | - Soo Young Cho
- Clinical Genomic Analysis Branch, Research Institute, National Cancer Center, Goyang, South Korea
| | - June Hyuk Kim
- National Cancer Center Hospital, National Cancer Center, Goyang, South Korea
| | - Hyun Guy Kang
- National Cancer Center Hospital, National Cancer Center, Goyang, South Korea
| | - Tak Yun
- National Cancer Center Hospital, National Cancer Center, Goyang, South Korea
| | - Sung Eun Oh
- National Cancer Center Hospital, National Cancer Center, Goyang, South Korea
| | - Seog Yun Park
- National Cancer Center Hospital, National Cancer Center, Goyang, South Korea
| | - Hye Jin You
- Division of Translational Science, Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang, Gyeonggi, 10408, South Korea. .,Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy (NCC-GCSP), National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang, Gyeonggi, 10408, South Korea.
| |
Collapse
|
16
|
Smrke A, Wang Y, Simmons C. Update on systemic therapy for advanced soft-tissue sarcoma. ACTA ACUST UNITED AC 2020; 27:25-33. [PMID: 32174755 DOI: 10.3747/co.27.5475] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background Soft-tissue sarcoma (sts) represents a rare group of mesenchymal neoplasms comprising more than 50 heterogeneous subtypes. Great efforts have been made to increase the understanding of the treatment of advanced sts (unresectable or metastatic disease). We set out to determine whether outcomes for patients with advanced sts have improved over time and to assess the current evidence for systemic therapy. Methods In a scoping review, we evaluated the contemporary evidence for systemic treatment of advanced sts in adults (>18 years of age). Phase i, ii, and iii studies of systemic therapy for advanced sts published in the English language were included. After abstract and full-text review of seventy-seven studies, sixty-two trials met the inclusion criteria. Results The number of clinical trials conducted and published in advanced sts has increased over the last 30 years. Although median overall survival has increased, attempts at improving first-line therapy through dose intensification, doublet chemotherapy, or alternative backbones have not been successful. The optimal therapy beyond anthracyclines remains a challenge, especially given the heterogeneity that grouping multiple sts subtypes within clinical trials creates. However, increasing numbers of agents are being studied, and several studies had shown isolated benefit in progression-free or overall survival. Summary First-line systemic therapy with an anthracycline remains the standard of care for advanced sts. However, choice of subsequent therapy beyond anthracyclines remains challenging. Novel systemic therapies, use of molecular diagnostics to direct therapy, subtype-specific trials, and learnings from real-world retrospective data are all important for improving outcomes in patients with advanced sts.
Collapse
Affiliation(s)
- A Smrke
- BC Cancer-Vancouver Centre, Vancouver, BC
| | - Y Wang
- BC Cancer-Vancouver Centre, Vancouver, BC
| | - C Simmons
- BC Cancer-Vancouver Centre, Vancouver, BC
| |
Collapse
|
17
|
Junaid M, Akter Y, Afrose SS, Tania M, Khan MA. Apoptotic Cell Death: Important Cellular Process as Chemotherapeutic Target. DRUG TARGETS IN CELLULAR PROCESSES OF CANCER: FROM NONCLINICAL TO PRECLINICAL MODELS 2020:65-88. [DOI: 10.1007/978-981-15-7586-0_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
|
18
|
Wong SHM, Kong WY, Fang CM, Loh HS, Chuah LH, Abdullah S, Ngai SC. The TRAIL to cancer therapy: Hindrances and potential solutions. Crit Rev Oncol Hematol 2019; 143:81-94. [PMID: 31561055 DOI: 10.1016/j.critrevonc.2019.08.008] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 12/15/2022] Open
Abstract
Apoptosis is an ordered and orchestrated cellular process that occurs in physiological and pathological conditions. Resistance to apoptosis is a hallmark of virtually all malignancies. Despite being a cause of pathological conditions, apoptosis could be a promising target in cancer treatment. Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), also known as Apo-2 ligand (Apo2L), is a member of TNF cytokine superfamily. It is a potent anti-cancer agent owing to its specific targeting towards cancerous cells, while sparing normal cells, to induce apoptosis. However, resistance occurs either intrinsically or after multiple treatments which may explain why cancer therapy fails. This review summarizes the apoptotic mechanisms via extrinsic and intrinsic apoptotic pathways, as well as the apoptotic resistance mechanisms. It also reviews the current clinically tested recombinant human TRAIL (rhTRAIL) and TRAIL receptor agonists (TRAs) against TRAIL-Receptors, TRAIL-R1 and TRAIL-R2, in which the outcomes of the clinical trials have not been satisfactory. Finally, this review discusses the current strategies in overcoming resistance to TRAIL-induced apoptosis in pre-clinical and clinical settings.
Collapse
Affiliation(s)
- Sonia How Ming Wong
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia
| | - Wei Yang Kong
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia
| | - Chee-Mun Fang
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia
| | - Hwei-San Loh
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia
| | - Lay-Hong Chuah
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia; Advanced Engineering Platform, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Syahril Abdullah
- Medical Genetics Laboratory, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, 43400 UPM, Malaysia; UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Selangor, 43400 UPM, Malaysia
| | - Siew Ching Ngai
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia.
| |
Collapse
|
19
|
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily that can initiate the apoptosis pathway by binding to its associated death receptors DR4 and DR5. The activation of the TRAIL pathway in inducing tumor-selective apoptosis leads to the development of TRAIL-based cancer therapies, which include recombinant forms of TRAIL, TRAIL receptor agonists, and other therapeutic agents. Importantly, TRAIL, DR4, and DR5 can all be induced by synthetic and natural agents that activate the TRAIL apoptosis pathway in cancer cells. Thus, understanding the regulation of the TRAIL apoptosis pathway can aid in the development of TRAIL-based therapies for the treatment of human cancer.
Collapse
|
20
|
Tanaka K, Kawano M, Iwasaki T, Itonaga I, Tsumura H. Surrogacy of intermediate endpoints for overall survival in randomized controlled trials of first-line treatment for advanced soft tissue sarcoma in the pre- and post-pazopanib era: a meta-analytic evaluation. BMC Cancer 2019; 19:56. [PMID: 30634944 PMCID: PMC6330427 DOI: 10.1186/s12885-019-5268-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 01/02/2019] [Indexed: 01/04/2023] Open
Abstract
Background Overall survival is the true endpoint for most randomized controlled trials (RCTs) of malignant tumors, whereas progression-free survival (PFS) is considered the most reliable surrogate endpoint for overall survival (OS). The present study aimed to evaluate the correlation between surrogate endpoints and OS in randomized trials of first-line chemotherapy with doxorubicin (DOX), the standard treatment for advanced and metastatic soft tissue sarcomas (ASTS), using a meta-analytic approach. Methods In a systematic review, we identified RCTs of first-line chemotherapy for ASTS that compared single-agent doxorubicin (DOX) with other chemotherapy regimens, and were published in English during January 1974–December 2017. A meta-analysis was performed to evaluate the efficacy of first-line treatments for ASTS. Surrogacy of the intermediate endpoints for OS was investigated using weighted linear regression analysis. Correlation strength was examined using the coefficient of determination (R2). Results Twenty-seven randomized trials, comprising 6156 patients (3371 patients in the experimental arm and 2785 patients in the DOX arm) were identified. The hazard ratios for OS and PFS showed that the efficacy of treatment for ASTS was not significantly different between standard DOX and experimental treatments. The median OS was significantly prolonged in RCTs published after 2012 when pazopanib was approved for treating ASTS. The median PFS, however, did not differ significantly. The correlation between PFS and OS was moderate (R2 = 0.557), but better than that between OS and 3-month PFS, 6-month PFS, and response rate (R2 = 0.200, 0.073, and 0.278, respectively). The correlation between PFS and OS tended to be more favorable in RCTs published after 2012 (R2 = 0.586 and 0.459, respectively). Conclusions The trial-level correlation between PFS and OS was only modest; it tended to be better in RCTs published after 2012. While the effective lines of chemotherapy and the introduction of new drugs prolonged OS but not PFS, PFS is a better surrogate than other intermediate endpoints in the first-line ASTS trials even in the post-pazopanib era. Although this does not negate the need for more reliable surrogate endpoints for OS. Electronic supplementary material The online version of this article (10.1186/s12885-019-5268-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kazuhiro Tanaka
- Department of Orthopaedic Surgery, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama, Yufu City, Oita, 879-5593, Japan.
| | - Masanori Kawano
- Department of Orthopaedic Surgery, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama, Yufu City, Oita, 879-5593, Japan
| | - Tatsuya Iwasaki
- Department of Orthopaedic Surgery, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama, Yufu City, Oita, 879-5593, Japan
| | - Ichiro Itonaga
- Department of Orthopaedic Surgery, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama, Yufu City, Oita, 879-5593, Japan
| | - Hiroshi Tsumura
- Department of Orthopaedic Surgery, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama, Yufu City, Oita, 879-5593, Japan
| |
Collapse
|
21
|
A meta-analysis of randomized controlled trials that compare standard doxorubicin with other first-line chemotherapies for advanced/metastatic soft tissue sarcomas. PLoS One 2019; 14:e0210671. [PMID: 30629708 PMCID: PMC6328231 DOI: 10.1371/journal.pone.0210671] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 12/28/2018] [Indexed: 12/14/2022] Open
Abstract
Objective The standard treatment for patients with advanced/metastatic soft tissue sarcomas (ASTS) is systemic chemotherapy with doxorubicin. A previous meta-analysis of 8 randomized controlled trials (RCTs) demonstrated the superiority of single-agent doxorubicin over doxorubicin-based combination chemotherapy for ASTS. However, meta-analyses of all RCTs that compare doxorubicin to other single-agent or combination regimens as first-line treatments for ASTS are lacking. We conducted a systematic review and meta-analysis to evaluate the efficacy and toxicity of current primary treatments for ASTS. Methods Eligible studies were RCTs of first-line chemotherapies for ASTS comparing doxorubicin alone to other single agents or to combination therapies (experimental arm). Data from studies reporting hazard ratios (HR) and 95% confidence intervals (CI) for overall survival (OS) and progression-free survival (PFS) were pooled. Other time-to-event endpoints were extracted from the studies based on Kaplan-Meier estimates, and pooled odds ratios (OR) and 95% CI were calculated. Results Twenty-seven eligible RCTs comprising 6156 patients were identified. Overall, the 1-year OS (OR 0.88, 95% CI 0.79–0.99, P = 0.03) was significantly improved in the experimental arm over the doxorubicin-only arm; however, there was no significant difference in 2-year OS (OR 0.87, 95% CI 0.73–1.03, P = 0.11) or OS (HR 0.97, 95% CI 0.91–1.03, P = 0.28) between the two groups. PFS and other time-to-event endpoints were not significantly different between the two treatment arms. While incidences of overall severe adverse events were not significantly different (OR 1.20, 95% CI 0.88–1.65, P = 0.26), severe nausea/vomiting was significantly more frequent in the experimental arm (OR 1.90, 95% CI 1.27–2.83, P = 0.002). Conclusion The efficacies of doxorubicin-only and experimental arm regimens were similar, although toxicities were more frequent in the experimental arms. Hence, doxorubicin monotherapy remains suitable as a standard first-line regimen for ASTS.
Collapse
|
22
|
Alemany R, Moura DS, Redondo A, Martinez-Trufero J, Calabuig S, Saus C, Obrador-Hevia A, Ramos R, Villar VH, Valverde C, Vaz MA, Medina J, Felipe-Abrio I, Hindi N, Taron M, Martin-Broto J. Nilotinib as Coadjuvant Treatment with Doxorubicin in Patients with Sarcomas: A Phase I Trial of the Spanish Group for Research on Sarcoma. Clin Cancer Res 2018; 24:5239-5249. [PMID: 30037815 DOI: 10.1158/1078-0432.ccr-18-0851] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/16/2018] [Accepted: 07/17/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Nilotinib plus doxorubicin showed to be synergistic regarding apoptosis in several sarcoma cell lines. A phase I/II trial was thus designed to explore the feasibility of nilotinib as coadjuvant of doxorubicin by inhibiting MRP-1/P-gp efflux activity. The phase I part of the study is presented here.Patients and Methods: Nilotinib 400 mg/12 hours was administered in fixed dose from day 1 to 6, and doxorubicin on day 5 of each cycle. Three dose escalation levels for doxorubicin at 60, 65, and 75 mg/m2 were planned. Cycles were repeated every 3 weeks for a total of 4 cycles. Eligible subtypes were retroperitoneal liposarcoma, leiomyosarcoma, and unresectable/metastatic high-grade chondrosarcoma.Results: Thirteen patients were enrolled: 7 chondrosarcoma, 4 liposarcoma, and 2 leiomyosarcoma. In 46 cycles administered, the most relevant grade 3/4 adverse effects per patient were neutropenia 54%, febrile neutropenia 15%, and asthenia 8%. No cardiac toxicity was observed. Only one dose-limiting toxicity (febrile neutropenia) was reported in the third dose level. With regard to efficacy, 1 partial response (1 liposarcoma), 9 stable diseases (5 chondrosarcoma, 2 liposarcoma, 1 leiomyosarcoma), and 3 progressive diseases (2 chondrosarcoma and 1 leiomyosarcoma) were present. ABCB1 and ABCC1 RNA expression levels decreased by 58.47-fold and 1.47-fold, respectively, on day 5 of the cycle.Conclusions: Combination of MRP-1/P-gp inhibitor, nilotinib, as coadjuvant with doxorubicin is feasible; it appears not to add substantial toxicity compared with doxorubicin alone. Pharmacodynamic study supports this concept. The recommended dose for the phase II part for doxorubicin was 75 mg/m2 Clin Cancer Res; 24(21); 5239-49. ©2018 AACR.
Collapse
Affiliation(s)
- Regina Alemany
- Department of Biology, Balearic Islands University, Palma de Mallorca, Spain.,Group of Advanced Therapies and Biomarkers in Clinical Oncology, Institut d'Investigació Sanitària de les Illes Balears (IdISBa-IUNICS), Palma de Mallorca, Spain
| | - David S Moura
- Instituto de Biomedicina de Sevilla (IBIS, HUVR, CSIC, Universidad de Sevilla), Lab.215, Sevilla, Spain
| | - Andres Redondo
- Medical Oncology Department, University Hospital La Paz, Madrid, Spain
| | | | - Silvia Calabuig
- Molecular Oncology Laboratory, Fundación Investigación, Hospital General Universitario de Valencia, Valencia, (Spain). Centro de Investigación Biomédica en Red de Cáncer (CIBEROnc), Madrid, Spain. Department of Pathology, Universitat de València, Valencia, Spain
| | - Carlos Saus
- Pathology Department, University Hospital Son Espases, Palma de Mallorca, Spain
| | - Antonia Obrador-Hevia
- Group of Advanced Therapies and Biomarkers in Clinical Oncology, Institut d'Investigació Sanitària de les Illes Balears (IdISBa-IUNICS), Palma de Mallorca, Spain.,Sequencing Unit, University Hospital Son Espases, Palma de Mallorca, Spain
| | - Rafael Ramos
- Pathology Department, University Hospital Son Espases, Palma de Mallorca, Spain
| | - Victor H Villar
- Department of Biology, Balearic Islands University, Palma de Mallorca, Spain
| | - Claudia Valverde
- Medical Oncology Department, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Maria Angeles Vaz
- Medical Oncology Department, University Hospital Ramon y Cajal, Madrid, Spain
| | - Javier Medina
- Medical Oncology Department, Hospital Virgen de la Salud, Toledo, Spain
| | - Irene Felipe-Abrio
- Instituto de Biomedicina de Sevilla (IBIS, HUVR, CSIC, Universidad de Sevilla), Lab.215, Sevilla, Spain.,Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre-CNIO, Madrid, Spain
| | - Nadia Hindi
- Instituto de Biomedicina de Sevilla (IBIS, HUVR, CSIC, Universidad de Sevilla), Lab.215, Sevilla, Spain.,Medical Oncology Department, University Hospital Virgen del Rocio, Sevilla, Spain
| | - Miguel Taron
- Instituto de Biomedicina de Sevilla (IBIS, HUVR, CSIC, Universidad de Sevilla), Lab.215, Sevilla, Spain
| | - Javier Martin-Broto
- Instituto de Biomedicina de Sevilla (IBIS, HUVR, CSIC, Universidad de Sevilla), Lab.215, Sevilla, Spain. .,Medical Oncology Department, University Hospital Virgen del Rocio, Sevilla, Spain
| |
Collapse
|
23
|
Multi-agent chemotherapy in advanced soft tissue sarcoma (STS) - A systematic review and meta-analysis. Cancer Treat Rev 2017; 63:71-78. [PMID: 29253836 DOI: 10.1016/j.ctrv.2017.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/02/2017] [Accepted: 12/05/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Despite a lack of improvement in overall survival (OS) with doxorubicin-based combinations over doxorubicin alone in advanced STS, the role of multi-agent chemotherapy remains poorly defined. METHODS We conducted a systematic review and meta-analysis to evaluate benefits and harms of multi-agent chemotherapy in advanced STS. Eligible studies were randomized trials of chemotherapy in advanced STS comparing single agent to multi-agent therapy. Data from studies reporting a hazard ratio (HR) and 95% confidence intervals (CI) for OS and progression-free survival (PFS) were pooled in a meta-analysis. Meta-regression was utilized to explore the association between efficacy (OS and PFS) and both toxicity and dose intensity. RESULTS We identified 22 trials published between 1974 and April 2016 and comprising 5044 patients. Overall, multi-agent chemotherapy was associated with improved OS (HR:0.79, p = 0.02), and borderline improvement in PFS (HR:0.86, p = 0.05). While the effect on OS was similar in trials with non-anthracycline controls compared to those with anthracycline controls (HR for OS 0.73 vs. 0.82, p for difference = 0.63) there was a non-significantly greater effect for multi-agent chemotherapy on PFS in non-anthracycline RCT (HR for PFS 0.73 vs. 0.91, p for difference = 0.13). Compared to studies with cytotoxic therapy-based multi-agent therapy, a non-significantly greater magnitude of effect among studies with biological/cytostatic experimental groups was seen (HR for OS 0.64 vs. 0.86, p for difference = 0.37). There was a borderline significant association between dose reductions (which were more common in combination arms) and worse PFS (beta = 0.70, p = 0.053). CONCLUSION Multi-agent chemotherapy is associated with a modest, but statistically significant improvement in outcomes in STS. Combining chemotherapy with non-cytotoxic agents might represent a promising strategy.
Collapse
|
24
|
Gamie Z, Kapriniotis K, Papanikolaou D, Haagensen E, Da Conceicao Ribeiro R, Dalgarno K, Krippner-Heidenreich A, Gerrand C, Tsiridis E, Rankin KS. TNF-related apoptosis-inducing ligand (TRAIL) for bone sarcoma treatment: Pre-clinical and clinical data. Cancer Lett 2017; 409:66-80. [PMID: 28888998 DOI: 10.1016/j.canlet.2017.08.036] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/24/2017] [Accepted: 08/28/2017] [Indexed: 01/25/2023]
Abstract
Bone sarcomas are rare, highly malignant mesenchymal tumours that affect teenagers and young adults, as well as older patients. Despite intensive, multimodal therapy, patients with bone sarcomas have poor 5-year survival, close to 50%, with lack of improvement over recent decades. TNF-related apoptosis-inducing ligand (TRAIL), a member of the tumour necrosis factor (TNF) ligand superfamily (TNFLSF), has been found to induce apoptosis in cancer cells while sparing nontransformed cells, and may therefore offer a promising new approach to treatment. We cover the existing preclinical and clinical evidence about the use of TRAIL and other death receptor agonists in bone sarcoma treatment. In vitro studies indicate that TRAIL and other death receptor agonists are generally potent against bone sarcoma cell lines. Ewing's sarcoma cell lines present the highest sensitivity, whereas osteosarcoma and chondrosarcoma cell lines are considered less sensitive. In vivo studies also demonstrate satisfactory results, especially in Ewing's sarcoma xenograft models. However, the few clinical trials in the literature show only low or moderate efficacy of TRAIL in treating bone sarcoma. Potential strategies to overcome the in vivo resistance reported include co-administration with other drugs and the potential to deliver TRAIL on the surface of primed mesenchymal or immune cells and the use of targeted single chain antibodies such as scFv-scTRAIL.
Collapse
Affiliation(s)
- Zakareya Gamie
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
| | - Konstantinos Kapriniotis
- Academic Orthopedic Department, "PapaGeorgiou" General Hospital, Thessaloniki, Greece; CORE-Center for Orthopedic Research at CIRI-AUTh, Aristotle University Medical School, Thessaloniki, Hellas, Greece.
| | - Dimitra Papanikolaou
- Academic Orthopedic Department, "PapaGeorgiou" General Hospital, Thessaloniki, Greece; CORE-Center for Orthopedic Research at CIRI-AUTh, Aristotle University Medical School, Thessaloniki, Hellas, Greece.
| | - Emma Haagensen
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
| | - Ricardo Da Conceicao Ribeiro
- School of Mechanical and Systems Engineering, Stephenson Building, Claremont Road, Newcastle Upon Tyne, NE1 7RU, UK.
| | - Kenneth Dalgarno
- School of Mechanical and Systems Engineering, Stephenson Building, Claremont Road, Newcastle Upon Tyne, NE1 7RU, UK.
| | - Anja Krippner-Heidenreich
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
| | - Craig Gerrand
- North of England Bone and Soft Tissue Tumour Service, Freeman Hospital, Freeman Road, Newcastle Upon Tyne, NE7 7DN, UK.
| | - Eleftherios Tsiridis
- Academic Orthopedic Department, "PapaGeorgiou" General Hospital, Thessaloniki, Greece; CORE-Center for Orthopedic Research at CIRI-AUTh, Aristotle University Medical School, Thessaloniki, Hellas, Greece; Secretary General European Hip Society, Austria.
| | - Kenneth Samora Rankin
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
| |
Collapse
|
25
|
Dominguez GA, Condamine T, Mony S, Hashimoto A, Wang F, Liu Q, Forero A, Bendell J, Witt R, Hockstein N, Kumar P, Gabrilovich DI. Selective Targeting of Myeloid-Derived Suppressor Cells in Cancer Patients Using DS-8273a, an Agonistic TRAIL-R2 Antibody. Clin Cancer Res 2016; 23:2942-2950. [PMID: 27965309 DOI: 10.1158/1078-0432.ccr-16-1784] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 11/11/2016] [Accepted: 12/02/2016] [Indexed: 01/01/2023]
Abstract
Purpose: Myeloid-derived suppressor cells (MDSC) are one of the major contributors to immune suppression in cancer. We recently have demonstrated in preclinical study that MDSCs are sensitive to TRAIL receptor 2 (TRAIL-R2) agonist. The goal of this study was to clinically test the hypothesis that targeting TRAIL-R2 can selectively eliminate MDSCs.Experimental Design: The TRAIL-R2 agonistic antibody (DS-8273a) has been tested in 16 patients with advanced cancers enrolled in a phase I trial. The antibody (24 mg/kg) was administered intravenously once every 3 weeks till disease progression, unacceptable toxicities, or withdrawal of consent. The safety and the presence of various populations of myeloid and lymphoid cells in peripheral blood and tumor tissues were evaluated.Results: The treatment was well tolerated with only mild to moderate adverse events attributable to the study drug. Treatment with DS-8273a resulted in reduction of the elevated numbers of MDSCs in the peripheral blood of most patients to the levels observed in healthy volunteers. However, in several patients, MDSCs rebounded back to the pretreatment level by day 42. In contrast, DS-8273a did not affect the number of neutrophils, monocytes, and other populations of myeloid and lymphoid cells. Decrease in MDSCs inversely correlated with the length of progression-free survival. In tumors, DS-8273a treatment resulted in a decrease of MDSCs in 50% of the patients who were able to provide pre- and on-treatment biopsies.Conclusions: Targeting TRAIL-R2 resulted in elimination of different populations of MDSCs without affecting mature myeloid or lymphoid cells. These data support the use of this antibody in combination immmunotherapy of cancer. Clin Cancer Res; 23(12); 2942-50. ©2016 AACR.
Collapse
Affiliation(s)
| | | | - Sridevi Mony
- The Wistar Institute, Philadelphia, Pennsylvania
| | | | - Fang Wang
- The Wistar Institute, Philadelphia, Pennsylvania
| | - Qin Liu
- The Wistar Institute, Philadelphia, Pennsylvania
| | - Andres Forero
- University of Alabama at Birmingham Comprehensive Cancer Center, Birmingham, Alabama
| | - Johanna Bendell
- Sarah Cannon Research Institute Tennessee Oncology, Nashville, Tennessee
| | - Robert Witt
- Helen F. Graham Cancer Center & Research Institute, Christiana Care Health System, Newark, Delaware
| | - Neil Hockstein
- Helen F. Graham Cancer Center & Research Institute, Christiana Care Health System, Newark, Delaware
| | | | | |
Collapse
|
26
|
Wang L, Chan JY, Zhou X, Cui G, Yan Z, Wang L, Yan R, Di L, Wang Y, Hoi MP, Shan L, Lee SM. A Novel Agent Enhances the Chemotherapeutic Efficacy of Doxorubicin in MCF-7 Breast Cancer Cells. Front Pharmacol 2016; 7:249. [PMID: 27559313 PMCID: PMC4979254 DOI: 10.3389/fphar.2016.00249] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 07/27/2016] [Indexed: 12/20/2022] Open
Abstract
We have previously demonstrated that DT-010, a novel conjugate of danshensu (DSS) and tetramethylpyrazine (TMP), displays anti-tumor effects in breast cancer cells both in vitro and in vivo. In the present study, we investigated whether DT-010 enhances the chemotherapeutic effect of doxorubicin (Dox) in MCF-7 breast cancer cells and exerts concurrent cardioprotective benefit at the same time. Our findings showed that DT-010 was more potent than TMP, DSS, or their combination in potentiating Dox-induced toxicity in MCF-7 cells. Co-treatment with DT-010 and Dox increased apoptosis in MCF-7 cells relative to Dox alone. Further study indicated that glycolytic capacity, glycolytic reserve and lactate level of MCF-7 cells were significantly inhibited after DT-010 treatment. DT-010 also increased the expression of the pro-survival protein GRP78, which was inhibited by co-treatment with Dox. Both endoplasmic reticulum stress inhibitor 4-PBA and knockdown of the expression of GRP78 protein potentiated DT-010-mediated apoptosis in MCF-7 cells. Moreover, DT-010 inhibited Dox-induced cardiotoxicity in H9c2 myoblasts. In conclusion, DT-010 and Dox confer synergistic anti-tumor effect in MCF-7 breast cancer cells through downregulation of the glycolytic pathway and inhibition of the expression of GRP78. Meanwhile, DT-010 also protects against Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Liang Wang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau Macao, China
| | - Judy Y Chan
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau Macao, China
| | - Xinhua Zhou
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau Macao, China
| | - Guozhen Cui
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau Macao, China
| | - Zhixiang Yan
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau Macao, China
| | - Li Wang
- Faculty of Health sciences, University of Macau Macao, China
| | - Ru Yan
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau Macao, China
| | - Lijun Di
- Faculty of Health sciences, University of Macau Macao, China
| | - Yuqiang Wang
- Institute of New Drug Research, College of Pharmacy, Jinan University Guangzhou, China
| | - Maggie P Hoi
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau Macao, China
| | - Luchen Shan
- Institute of New Drug Research, College of Pharmacy, Jinan University Guangzhou, China
| | - Simon M Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau Macao, China
| |
Collapse
|
27
|
Novel Water-Borne Polyurethane Nanomicelles for Cancer Chemotherapy: Higher Efficiency of Folate Receptors Than TRAIL Receptors in a Cancerous Balb/C Mouse Model. Pharm Res 2016; 33:1426-39. [PMID: 26908046 DOI: 10.1007/s11095-016-1884-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/16/2016] [Indexed: 01/17/2023]
Abstract
PURPOSE Since the introduction of nanocarriers, the delivery of chemotherapeutic agents for treatment of patients with cancer has been possible with better effectiveness. The latest findings are also support that further enhancement in therapeutic effectiveness of these nanocarriers can be attained, if surface decoration with proper targeting agents is considered. METHODS This study aimed at treating a variety of 4T1 murine breast cancer cell line, mainly demonstrating high folate and TRAIL receptor expression of cancerous cells. The therapeutic efficacy of paclitaxel loaded Cremophore EL (Taxol®), paclitaxel loaded waterborne polyurethane nanomicelles (PTX-PU) and paclitaxel loaded waterborne polyurethane nanomicelles conjugated with folate (PTX-PU-FA) and TRAIL (PTX-PU-TRAIL) on treating 4T1 cell was also compared. RESULTS The findings that worth noting are: PTX-PU outperformed Taxol® in a Balb/C mouse model, furthermore, tumor growth was adequately curbed by folate and TRAIL-decorated nanomicelles rather than the unconjugated formulation. Tumors of mice treated with PTX-PU-FA and PTX-PU-TRAIL shrank substantially compared to those treated with Taxol®, PTX-PU and PTX-PU-TRAIL (average 573 mm(3) versus 2640, 846, 717 mm(3) respectively), 45 days subsequent to tumor inoculation. The microscopic study of hematoxylin-eosin stained tumors tissue and apoptotic cell fraction substantiated that the most successful therapeutic effects have been observed for the mice treated with PTX-PU-FA (about 90% in PTX-PU-FA versus 75%, 60%, 15% in PTX-PU-TRAIL, PTX-PU, and Taxol® group respectively). CONCLUSIONS Using folate-targeted nanocarriers to treat cancers characterized by a high level of folate ligand expression is well substantiated by the findings of this study.
Collapse
|
28
|
Kang Z, Goldstein SD, Yu Y, Meltzer PS, Loeb DM, Cao L. Caspase-8 expression is predictive of tumour response to death receptor 5 agonist antibody in Ewing's sarcoma. Br J Cancer 2015; 113:894-901. [PMID: 26291055 PMCID: PMC4578089 DOI: 10.1038/bjc.2015.298] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 07/08/2015] [Accepted: 07/22/2015] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Despite good initial response to chemotherapy, 30% of Ewing's sarcoma (EWS) patients with localised tumours develop recurrent disease, associated with poor prognosis. The aim of this study was to address this challenge by conducting preclinical evaluation of a death receptor targeted agent in vitro and in vivo, and by identifying predictive biomarkers. METHODS Cell viability assays, drug dose responses, immunoblots, rescue with gene transfer, mice tumour models, and statistical comparisons of tumour growth and Kaplan-Meier survival curves. RESULTS This study shows that many EWS cell lines are selectively sensitive to a death receptor DR5 antibody and are more resistant to a DR4 antibody. Preclinical evaluation of these cell lines indicates their sensitivity to human DR5 agonist antibody conatumumab in vitro, which induces rapid activation of caspase-8 and apoptosis. We also found that sensitivity to conatumumab correlates with expression of caspase-8. Furthermore, the catalytic activity of caspase-8 is both necessary and sufficient to confer this sensitivity. In vivo, conatumumab is active against an EWS cell line and a patient-derived xenograft with higher caspase-8 expression, but is not effective against another with lower caspase-8 expression. CONCLUSIONS These studies suggest the potential of conatumumab as a therapeutic agent against EWS and caspase-8 as a predictive biomarker for sensitivity.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Apoptosis
- Biomarkers, Tumor/immunology
- Biomarkers, Tumor/metabolism
- Bone Neoplasms/drug therapy
- Bone Neoplasms/enzymology
- Bone Neoplasms/immunology
- Caspase 8/metabolism
- Cell Line, Tumor
- Drug Resistance, Neoplasm
- Female
- Heterografts
- Humans
- Mice
- Random Allocation
- Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists
- Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology
- Sarcoma, Ewing/drug therapy
- Sarcoma, Ewing/enzymology
- Sarcoma, Ewing/immunology
- Xenograft Model Antitumor Assays/methods
Collapse
Affiliation(s)
- Zhigang Kang
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
- Basic Science Program, Leidos Biomedical Research, Inc, Frederick, MD 21702, USA
| | - Seth D Goldstein
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Yunkai Yu
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Paul S Meltzer
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - David M Loeb
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Liang Cao
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
29
|
Amarante-Mendes GP, Griffith TS. Therapeutic applications of TRAIL receptor agonists in cancer and beyond. Pharmacol Ther 2015; 155:117-31. [PMID: 26343199 DOI: 10.1016/j.pharmthera.2015.09.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
TRAIL/Apo-2L is a member of the TNF superfamily first described as an apoptosis-inducing cytokine in 1995. Similar to TNF and Fas ligand, TRAIL induces apoptosis in caspase-dependent manner following TRAIL death receptor trimerization. Because tumor cells were shown to be particularly sensitive to this cytokine while normal cells/tissues proved to be resistant along with being able to synthesize and release TRAIL, it was rapidly appreciated that TRAIL likely served as one of our major physiologic weapons against cancer. In line with this, a number of research laboratories and pharmaceutical companies have attempted to exploit the ability of TRAIL to kill cancer cells by developing recombinant forms of TRAIL or TRAIL receptor agonists (e.g., receptor-specific mAb) for therapeutic purposes. In this review article we will describe the biochemical pathways used by TRAIL to induce different cell death programs. We will also summarize the clinical trials related to this pathway and discuss possible novel uses of TRAIL-related therapies. In recent years, the physiological importance of TRAIL has expanded beyond being a tumoricidal molecule to one critical for a number of clinical settings - ranging from infectious disease and autoimmunity to cardiovascular anomalies. We will also highlight some of these conditions where modulation of the TRAIL/TRAIL receptor system may be targeted in the future.
Collapse
Affiliation(s)
- Gustavo P Amarante-Mendes
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, SP, Brazil; Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia, Brazil.
| | - Thomas S Griffith
- Department of Urology, Masonic Cancer Center, Center for Immunology, University of Minnesota, Minneapolis, MN, USA; Minneapolis VA Health Care System, Minneapolis, MN 55417, USA.
| |
Collapse
|
30
|
Ciprotti M, Tebbutt NC, Lee FT, Lee ST, Gan HK, McKee DC, O'Keefe GJ, Gong SJ, Chong G, Hopkins W, Chappell B, Scott FE, Brechbiel MW, Tse AN, Jansen M, Matsumura M, Kotsuma M, Watanabe R, Venhaus R, Beckman RA, Greenberg J, Scott AM. Phase I Imaging and Pharmacodynamic Trial of CS-1008 in Patients With Metastatic Colorectal Cancer. J Clin Oncol 2015; 33:2609-16. [PMID: 26124477 DOI: 10.1200/jco.2014.60.4256] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
PURPOSE CS-1008 (tigatuzumab) is a humanized, monoclonal immunoglobulin G1 (IgG1) agonistic antibody to human death receptor 5. The purpose of this study was to investigate the impact of CS-1008 dose on the biodistribution, quantitative tumor uptake, and antitumor response in patients with metastatic colorectal cancer (mCRC). PATIENTS AND METHODS Patients with mCRC who had received at least one course of chemotherapy were assigned to one of five dosage cohorts and infused with a weekly dose of CS-1008. Day 1 and day 36 doses were trace-labeled with indium-111 ((111)In), followed by whole-body planar and regional single-photon emission computed tomography (SPECT) imaging at several time points over the course of 10 days. RESULTS Nineteen patients were enrolled. (111)In-CS-1008 uptake in tumor was observed in only 12 patients (63%). (111)In-CS-1008 uptake and pharmacokinetics were not affected by dose or repeated drug administration. (111)In-CS-1008 biodistribution showed gradual blood-pool clearance and no abnormal uptake in normal tissue. No anti-CS-1008 antibody development was detected. One patient achieved partial response (3.7 months duration), eight patients had stable disease, and 10 patients had progressive disease. Clinical benefit rate (stable disease + partial response) in patients with (111)In-CS-1008 uptake in tumor was 58% versus 28% in patients with no uptake. An analysis of individual lesions showed that lesions with antibody uptake were one third as likely to progress as those without antibody uptake (P = .07). Death-receptor-5 expression in archived tumor samples did not correlate with (111)In-CS-1008 uptake (P = .5) or tumor response (P = .6). CONCLUSION Death-receptor-5 imaging with (111)In-CS-1008 reveals interpatient and intrapatient heterogeneity of uptake in tumor, is not dose dependent, and is predictive of clinical benefit in the treatment of patients who have mCRC.
Collapse
Affiliation(s)
- Marika Ciprotti
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Niall C Tebbutt
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Fook-Thean Lee
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Sze-Ting Lee
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Hui K Gan
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - David C McKee
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Graeme J O'Keefe
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Sylvia J Gong
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Geoffrey Chong
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Wendie Hopkins
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Bridget Chappell
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Fiona E Scott
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Martin W Brechbiel
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Archie N Tse
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Mendel Jansen
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Manabu Matsumura
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Masakatsu Kotsuma
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Rira Watanabe
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Ralph Venhaus
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Robert A Beckman
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Jonathan Greenberg
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Andrew M Scott
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY.
| |
Collapse
|
31
|
Lim B, Allen JE, Prabhu VV, Talekar MK, Finnberg NK, El-Deiry WS. Targeting TRAIL in the treatment of cancer: new developments. Expert Opin Ther Targets 2015; 19:1171-85. [DOI: 10.1517/14728222.2015.1049838] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
32
|
Dai X, Zhang J, Arfuso F, Chinnathambi A, Zayed ME, Alharbi SA, Kumar AP, Ahn KS, Sethi G. Targeting TNF-related apoptosis-inducing ligand (TRAIL) receptor by natural products as a potential therapeutic approach for cancer therapy. Exp Biol Med (Maywood) 2015; 240:760-73. [PMID: 25854879 DOI: 10.1177/1535370215579167] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been shown to selectively induce apoptotic cell death in various tumor cells by engaging its death-inducing receptors (TRAIL-R1 and TRAIL-R2). This property has led to the development of a number of TRAIL-receptor agonists such as the soluble recombinant TRAIL and agonistic antibodies, which have shown promising anticancer activity in preclinical studies. However, besides activating caspase-dependent apoptosis in several cancer cells, TRAIL may also activate nonapoptotic signal transduction pathways such as nuclear factor-kappa B, mitogen-activated protein kinases, AKT, and signal transducers and activators of transcription 3, which may contribute to TRAIL resistance that is being now frequently encountered in various cancers. TRAIL resistance can be overcome by the application of efficient TRAIL-sensitizing pharmacological agents. Natural compounds have shown a great potential in sensitizing cells to TRAIL treatment through suppression of distinct survival pathways. In this review, we have summarized both apoptotic and nonapoptotic pathways activated by TRAIL, as well as recent advances in developing TRAIL-receptor agonists for cancer therapy. We also briefly discuss combination therapies that have shown great potential in overcoming TRAIL resistance in various tumors.
Collapse
Affiliation(s)
- Xiaoyun Dai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Jingwen Zhang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Frank Arfuso
- School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Curtin University, Western Australia 6009, Australia
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - M E Zayed
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Curtin University, Western Australia 6009, Australia Cancer Science Institute of Singapore, Centre for Translational Medicine, Singapore 117599, Singapore Department of Biological Sciences, University of North Texas, Denton, TX 76203, USA
| | - Kwang Seok Ahn
- College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Curtin University, Western Australia 6009, Australia Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| |
Collapse
|
33
|
Arif IS, Hooper CL, Greco F, Williams AC, Boateng SY. Increasing doxorubicin activity against breast cancer cells using PPARγ-ligands and by exploiting circadian rhythms. Br J Pharmacol 2015; 169:1178-88. [PMID: 23578093 DOI: 10.1111/bph.12202] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 01/25/2013] [Accepted: 04/09/2013] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND PURPOSE Doxorubicin is effective against breast cancer, but its major side effect is cardiotoxicity. The aim of this study was to determine whether the efficacy of doxorubicin on cancer cells could be increased in combination with PPARγ agonists or chrono-optimization by exploiting the diurnal cycle. EXPERIMENTAL APPROACH We determined cell toxicity using MCF-7 cancer cells, neonatal rat cardiac myocytes and fibroblasts in this study. KEY RESULTS Doxorubicin damages the contractile filaments of cardiac myocytes and affects cardiac fibroblasts by significantly inhibiting collagen production and proliferation at the level of the cell cycle. Cyclin D1 protein levels decreased significantly following doxorubicin treatment indicative of a G1/S arrest. PPARγ agonists with doxorubicin increased the toxicity to MCF-7 cancer cells without affecting cardiac cells. Rosiglitazone and ciglitazone both enhanced anti-cancer activity when combined with doxorubicin (e.g. 50% cell death for doxorubicin at 0.1 μM compared to 80% cell death when combined with rosiglitazone). Thus, the therapeutic dose of doxorubicin could be reduced by 20-fold through combination with the PPARγ agonists, thereby reducing adverse effects on the heart. The presence of melatonin also significantly increased doxorubicin toxicity, in cardiac fibroblasts (1 μM melatonin) but not in MCF-7 cells. CONCLUSIONS AND IMPLICATIONS Our data show, for the first time, that circadian rhythms play an important role in doxorubicin toxicity in the myocardium; doxorubicin should be administered mid-morning, when circulating levels of melatonin are low, and in combination with rosiglitazone to increase therapeutic efficacy in cancer cells while reducing the toxic effects on the heart.
Collapse
Affiliation(s)
- I S Arif
- School of Pharmacy, University of Reading, Reading, UK
| | | | | | | | | |
Collapse
|
34
|
Unexpected hepatotoxicity in a phase I study of TAS266, a novel tetravalent agonistic Nanobody® targeting the DR5 receptor. Cancer Chemother Pharmacol 2015; 75:887-95. [DOI: 10.1007/s00280-015-2712-0] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/19/2015] [Indexed: 11/26/2022]
|
35
|
Redjal N, Zhu Y, Shah K. Combination of systemic chemotherapy with local stem cell delivered S-TRAIL in resected brain tumors. Stem Cells 2015; 33:101-10. [PMID: 25186100 PMCID: PMC4270944 DOI: 10.1002/stem.1834] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 07/29/2014] [Indexed: 01/02/2023]
Abstract
Despite advances in standard therapies, the survival of glioblastoma multiforme (GBM) patients has not improved. Limitations to successful translation of new therapies include poor delivery of systemic therapies and use of simplified preclinical models which fail to reflect the clinical complexity of GBMs. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces apoptosis specifically in tumor cells and we have tested its efficacy by on-site delivery via engineered stem cells (SC) in mouse models of GBM that mimic the clinical scenario of tumor aggressiveness and resection. However, about half of tumor lines are resistant to TRAIL and overcoming TRAIL-resistance in GBM by combining therapeutic agents that are currently in clinical trials with SC-TRAIL and understanding the molecular dynamics of these combination therapies are critical to the broad use of TRAIL as a therapeutic agent in clinics. In this study, we screened clinically relevant chemotherapeutic agents for their ability to sensitize resistant GBM cell lines to TRAIL induced apoptosis. We show that low dose cisplatin increases surface receptor expression of death receptor 4/5 post G2 cycle arrest and sensitizes GBM cells to TRAIL induced apoptosis. In vivo, using an intracranial resection model of resistant primary human-derived GBM and real-time optical imaging, we show that a low dose of cisplatin in combination with synthetic extracellular matrix encapsulated SC-TRAIL significantly decreases tumor regrowth and increases survival in mice bearing GBM. This study has the potential to help expedite effective translation of local stem cell-based delivery of TRAIL into the clinical setting to target a broad spectrum of GBMs.
Collapse
Affiliation(s)
- Navid Redjal
- Molecular Neurotherapy and Imaging Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Yanni Zhu
- Molecular Neurotherapy and Imaging Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Khalid Shah
- Molecular Neurotherapy and Imaging Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
| |
Collapse
|
36
|
Rocchi L, Caraffi S, Perris R, Mangieri D. The angiogenic asset of soft tissue sarcomas: a new tool to discover new therapeutic targets. Biosci Rep 2014; 34:e00147. [PMID: 25236925 PMCID: PMC4219423 DOI: 10.1042/bsr20140075] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 07/04/2014] [Accepted: 07/14/2014] [Indexed: 12/18/2022] Open
Abstract
STS (soft tissue sarcomas) are rare malignant tumours deriving from cells of mesenchymal origin and represent only 1% of all malignant neoplasms. It has been extensively demonstrated that angiogenesis has an important role in cancer malignancy. Particularly, a lot of studies demonstrate the importance of angiogenesis in the development of carcinomas, whereas little is known about the role of angiogenesis in sarcomas and especially in STS. This review aims at summarizing the new discoveries about the nature and the importance of angiogenesis in STS and the new possible therapeutic strategies involved. Only a few studies concerning STS focus on tumour neovascularization and proangiogenic factors and look for a correlation with the patients prognosis/survival. These studies demonstrate that intratumoural MVD (microvessels density) may not accurately represent the angiogenic capacity of STS. Nevertheless, this does not exclude the possibility that angiogenesis could be important in STS. The importance of neoangiogenesis in soft tissue tumours is confirmed by the arising number of publications comparing angiogenesis mediators with clinical features of patients with STS. The efficacy of anti-angiogenic therapies in other types of cancer is well documented. The understanding of the involvement of the angiogenic process in STS, together with the necessity to improve the therapy for this often mortal condition, prompted the exploration of anti-tumour compounds targeting this pathway. In conclusion, this review emphasizes the importance to better understand the mechanisms of angiogenesis in STS in order to subsequently design-specific target therapies for this group of poorly responding tumours.
Collapse
Key Words
- angiogenesis factors
- angiogenesis
- soft tissue sarcomas
- target therapy
- csf, colony-stimulating factor
- ec, endothelial cell
- fgf-2, fibroblast growth factor-2
- mfh, malignant fibrous histiocytoma
- mmp, matrix metalloproteinase
- mtor, mammalian target of rapamycin
- mvd, microvessels density
- pdgfrβ, platelet-derived growth factor beta
- plgf, placental growth factor
- sts, soft tissue sarcomas
- tki, tyrosine kinase inhibitor
- timp, tissue inhibitors of metalloproteinases
- upa, urokinase-type plasminogen activator
- vegf, vascular endothelial growth factor
- vegfr, vegf receptor
- vwf, von-willebrand factor
Collapse
Affiliation(s)
- Laura Rocchi
- *Unità Operativa di Anatomia e Istologia Patologica, Azienda Ospedaliero-Universitaria di Parma, Via Gramsci, 14, 43100-Parma, Italy
| | - Stefano Caraffi
- *Unità Operativa di Anatomia e Istologia Patologica, Azienda Ospedaliero-Universitaria di Parma, Via Gramsci, 14, 43100-Parma, Italy
| | - Roberto Perris
- †COMT–Centro di Oncologia Medica e Traslazionale, Università di Parma, Parco Area delle Scienze 11/A 43100-Parma, Italy
| | - Domenica Mangieri
- *Unità Operativa di Anatomia e Istologia Patologica, Azienda Ospedaliero-Universitaria di Parma, Via Gramsci, 14, 43100-Parma, Italy
- †COMT–Centro di Oncologia Medica e Traslazionale, Università di Parma, Parco Area delle Scienze 11/A 43100-Parma, Italy
| |
Collapse
|
37
|
Lemke J, von Karstedt S, Zinngrebe J, Walczak H. Getting TRAIL back on track for cancer therapy. Cell Death Differ 2014; 21:1350-64. [PMID: 24948009 PMCID: PMC4131183 DOI: 10.1038/cdd.2014.81] [Citation(s) in RCA: 377] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/14/2014] [Accepted: 04/15/2014] [Indexed: 02/07/2023] Open
Abstract
Unlike other members of the TNF superfamily, the TNF-related apoptosis-inducing ligand (TRAIL, also known as Apo2L) possesses the unique capacity to induce apoptosis selectively in cancer cells in vitro and in vivo. This exciting discovery provided the basis for the development of TRAIL-receptor agonists (TRAs), which have demonstrated robust anticancer activity in a number of preclinical studies. Subsequently initiated clinical trials testing TRAs demonstrated, on the one hand, broad tolerability but revealed, on the other, that therapeutic benefit was rather limited. Several factors that are likely to account for TRAs' sobering clinical performance have since been identified. First, because of initial concerns over potential hepatotoxicity, TRAs with relatively weak agonistic activity were selected to enter clinical trials. Second, although TRAIL can induce apoptosis in several cancer cell lines, it has now emerged that many others, and importantly, most primary cancer cells are resistant to TRAIL monotherapy. Third, so far patients enrolled in TRA-employing clinical trials were not selected for likelihood of benefitting from a TRA-comprising therapy on the basis of a valid(ated) biomarker. This review summarizes and discusses the results achieved so far in TRA-employing clinical trials in the light of these three shortcomings. By integrating recent insight on apoptotic and non-apoptotic TRAIL signaling in cancer cells, we propose approaches to introduce novel, revised TRAIL-based therapeutic concepts into the cancer clinic. These include (i) the use of recently developed highly active TRAs, (ii) the addition of efficient, but cancer-cell-selective TRAIL-sensitizing agents to overcome TRAIL resistance and (iii) employing proteomic profiling to uncover resistance mechanisms. We envisage that this shall enable the design of effective TRA-comprising therapeutic concepts for individual cancer patients in the future.
Collapse
Affiliation(s)
- J Lemke
- 1] Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK [2] Clinic of General and Visceral Surgery, University of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - S von Karstedt
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - J Zinngrebe
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - H Walczak
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| |
Collapse
|
38
|
Cancer therapeutics: Targeting the apoptotic pathway. Crit Rev Oncol Hematol 2014; 90:200-19. [DOI: 10.1016/j.critrevonc.2013.12.012] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Revised: 12/05/2013] [Accepted: 12/12/2013] [Indexed: 01/20/2023] Open
|
39
|
Sun S, Li Z, Sun L, Yang C, Mei Z, Ouyang W, Yang B, Xie C. Results on efficacy and safety of cancer treatment with or without tumor necrosis factor-related apoptosis-inducing ligand-related agents: A meta-analysis. Mol Clin Oncol 2014; 2:440-448. [PMID: 24772315 DOI: 10.3892/mco.2014.261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 01/21/2014] [Indexed: 11/05/2022] Open
Abstract
This meta-analysis aimed to evaluate the currently available evidence on the efficacy and safety of cancer treatment with or without tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-related agents. We conducted a systematic search through Medline, Cochrane Library and EMBASE electronic databases and manually searched the Journal of Clinical Oncology to identify randomized controlled trials (RCTs) conducted between 1995 and 2013 comparing the efficacy and safety results of cancer treatment with and without TRAIL-related agents. The methodological quality of the included RCTs was assessed by the Cochrane Risk of Bias assessment tool. The outcome measurements included objective response rate (ORR), clinical benefit rate (CBR)/disease control rate (DCR) and adverse events (AEs). The relevant data were analyzed using Review Manager 5.2 software. Grading of Recommendations Assessment Development and Evaluation was used to assess the quality of evidence and grade of recommendation. Four RCTs, including a total of 596 patients, were ultimately selected and analyzed. There were no statistically significant differences among the 4 RCTs regarding ORR [relative risk (RR)=0.92, 95% confidence interval (CI): 0.73-1.15, P=0.45], CBR/DCR (RR=0.92, 95% CI: 0.81-1.05, P=0.21), progression-free survival [hazard ratio (HR)=0.89, 95% CI: 0.75-1.05, P=0.16], overall survival (HR=0.90, 95% CI: 0.74-1.09, P=0.27), number of patients with any AEs (RR=0.99, 95% CI: 0.96-1.03, P=0.77), number of patients with any severe AEs (RR=0.95, 95% CI: 0.78-1.55, P=0.58), number of patients with ≥grade 3 AEs (RR=1.13, 95% CI: 0.93-1.38, P=0.22) and number of fatal AEs (RR=1.14, 95% CI: 0.71-1.81, P=0.59). The quality of evidence was considered to be moderate and the recommendation grades were weak. In conclusion, the benefits of TRAIL-related agents in the treatment of cancer patients remain uncertain and further clinical trials are required.
Collapse
Affiliation(s)
- Shaoxing Sun
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zonghuan Li
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Li Sun
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Chunxu Yang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zijie Mei
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Wen Ouyang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Bo Yang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Conghua Xie
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China ; ; Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
40
|
Abstract
Apoptosis is a cell death program that is well-orchestrated for normal tissue homeostasis and for removal of damaged, old or infected cells. It is regulated by intrinsic and extrinsic pathways. The intrinsic pathway responds to signals such as ultraviolet radiation or DNA damage and activates "executioner" caspases through a mitochondria-dependent pathway. The extrinsic pathway is activated by death signals induced, for example, by an infection that activates the immune system or receptor-mediated pathways. The extrinsic pathway signals also cascade down to executioner caspases that cleave target proteins and lead to cell death. Strict control of cellular apoptosis is important for the hematopoietic system as it has a high turnover rate. However, the apoptosis program is often deregulated in hematologic malignancies leading to the accumulation of malignant cells. Therefore, apoptosis pathways have been identified for the development of anticancer therapeutics. We review here the proteins that have been targeted for anticancer drug development in hematologic malignancies. These include BCL-2 family proteins, death ligands and receptors, inhibitor of apoptosis family proteins and caspases. Except for caspase activators, drugs that target each of these classes of proteins have advanced into clinical trials.
Collapse
Affiliation(s)
- Shadia Zaman
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center , Houston, TX , USA
| | | | | |
Collapse
|
41
|
Holland PM. Death receptor agonist therapies for cancer, which is the right TRAIL? Cytokine Growth Factor Rev 2013; 25:185-93. [PMID: 24418173 DOI: 10.1016/j.cytogfr.2013.12.009] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 12/15/2013] [Indexed: 01/28/2023]
Abstract
The activation of cell-surface death receptors represents an attractive therapeutic strategy to promote apoptosis of tumor cells. Several investigational therapeutics that target this extrinsic pathway, including recombinant human Apo2L/TRAIL and monoclonal agonist antibodies directed against death receptors-4 (DR4) or -5 (DR5), have been evaluated in the clinic. Although Phase 1/1b studies provided encouraging preliminary results, findings from randomized Phase 2 studies failed to demonstrate significant clinical benefit. This has raised multiple questions as to why pre-clinical data were not predictive of clinical response. Results from clinical studies and insight into why current agents have failed to yield robust responses are discussed. In addition, new strategies for the development of next generation death receptor agonists are reviewed.
Collapse
Affiliation(s)
- Pamela M Holland
- Therapeutic Innovation Unit, Amgen Inc., 360 Binney Street, Cambridge, MA 02142, United States.
| |
Collapse
|
42
|
Ploner F, Lamm W, Schur S, Eisterer W, Kühr T, Lindorfer A, Tinchon C, Köstler WJ, Szkandera J, Brodowicz T. The Austrian experience with trabectedin in non-selected patients with metastatic soft tissue sarcoma (STS). J Cancer Res Clin Oncol 2013; 139:1337-42. [PMID: 23666164 DOI: 10.1007/s00432-013-1447-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 04/26/2013] [Indexed: 11/28/2022]
Abstract
PURPOSE The purpose of this retrospective analysis was to assess efficacy and tolerability of trabectedin in soft tissue sarcoma (STS) in the routine clinical setting. PATIENTS AND METHODS Efficacy and safety data of trabectedin were retrospectively evaluated in patients with advanced STS who had started treatment with trabectedin at six institutions in Austria between January 2008 and May 2012. RESULTS Data of 101 adult patients were included in the present analysis. Patients had a median age of 56 years; 59 and 41% received trabectedin as ≤2nd and ≥3rd chemotherapy line for advanced disease, respectively. Median progression-free survival (PFS) and overall survival (OS) were 3.9 and 11.6 months. Median PFS and OS were different in patients who received trabectedin as ≤2nd- or ≥3rd-line treatment: median PFS was 3.9 versus 3.6 months and OS was 15.2 versus 24.8 months, respectively. The extent and severity of trabectedin-induced toxicity were low and manageable. CONCLUSIONS The activity and tolerability of trabectedin in the routine clinical setting is comparable to outcomes reported in phase II trials already published. Regardless of whether trabectedin was given earlier or later in the course of disease, outcomes did not differ in the cohort of analysed patients.
Collapse
Affiliation(s)
- F Ploner
- Department of Oncology, Medical University Graz, Auenbruggerplatz 15, 8036 Graz, Austria.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Micheau O, Shirley S, Dufour F. Death receptors as targets in cancer. Br J Pharmacol 2013; 169:1723-44. [PMID: 23638798 PMCID: PMC3753832 DOI: 10.1111/bph.12238] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 04/25/2013] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Anti-tumour therapies based on the use pro-apoptotic receptor agonists, including TNF-related apoptosis-inducing ligand (TRAIL) or monoclonal antibodies targeting TRAIL-R1 or TRAIL-R2, have been disappointing so far, despite clear evidence of clinical activity and lack of adverse events for the vast majority of these compounds, whether combined or not with conventional or targeted anti-cancer therapies. This brief review aims at discussing the possible reasons for the lack of apparent success of these therapeutic approaches and at providing hints in order to rationally design optimal protocols based on our current understanding of TRAIL signalling regulation or resistance for future clinical trials. LINKED ARTICLES This article is part of a themed section on Emerging Therapeutic Aspects in Oncology. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2013.169.issue-8.
Collapse
|
44
|
Cohn AL, Tabernero J, Maurel J, Nowara E, Sastre J, Chuah BYS, Kopp MV, Sakaeva DD, Mitchell EP, Dubey S, Suzuki S, Hei YJ, Galimi F, McCaffery I, Pan Y, Loberg R, Cottrell S, Choo SP. A randomized, placebo-controlled phase 2 study of ganitumab or conatumumab in combination with FOLFIRI for second-line treatment of mutant KRAS metastatic colorectal cancer. Ann Oncol 2013; 24:1777-1785. [PMID: 23510984 DOI: 10.1093/annonc/mdt057] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Targeted agents presently available for mutant KRAS metastatic colorectal cancer (mCRC) are bevacizumab and aflibercept. We evaluated the efficacy and safety of conatumumab (an agonistic monoclonal antibody against human death receptor 5) and ganitumab (a monoclonal antibody against the type 1 insulin-like growth factor receptor) combined with standard FOLFIRI chemotherapy as a second-line treatment in patients with mutant KRAS mCRC. PATIENTS AND METHODS Patients with mutant KRAS metastatic adenocarcinoma of the colon or rectum refractory to fluoropyrimidine- and oxaliplatin-based chemotherapy were randomized 1 : 1 : 1 to receive intravenous FOLFIRI plus conatumumab 10 mg/kg (Arm A), ganitumab 12 mg/kg (Arm B), or placebo (Arm C) Q2W. The primary end point was progression-free survival (PFS). RESULTS In total, 155 patients were randomized. Median PFS in Arms A, B, and C was 6.5 months (HR, 0.69; P = 0.147), 4.5 months (HR, 1.01; P = 0.998), and 4.6 months, respectively; median overall survival was 12.3 months (HR, 0.89; P = 0.650), 12.4 months (HR, 1.27; P = 0.357), and 12.0 months; and objective response rate was 14%, 8%, and 2%. The most common grade ≥3 adverse events in Arms A/B/C included neutropenia (30%/25%/18%) and diarrhea (18%/2%/10%). CONCLUSIONS Conatumumab, but not ganitumab, plus FOLFIRI was associated with a trend toward improved PFS. Both combinations had acceptable toxicity.
Collapse
Affiliation(s)
- A L Cohn
- Rocky Mountain Cancer Center, Denver, USA.
| | - J Tabernero
- Medical Oncology Department, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona
| | - J Maurel
- Medical Oncology Department, Hospital Clinic de Barcelona, Barcelona, Spain
| | - E Nowara
- Maria Skodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - J Sastre
- Hospital Clinico San Carlos, Servicio de Oncologíca Medíca, Madrid, and Instituto Carlos III, Spanish Ministry of Science and Innovation, Madrid, Spain
| | - B Y S Chuah
- Department of Internal Medicine, National University Hospital, Singapore, Singapore
| | - M V Kopp
- Samara Regional Oncology Dispensary, Samara
| | - D D Sakaeva
- Clinical Oncology Dispensary of the Republic of Bashkortostan, Ufa, Russia
| | - E P Mitchell
- Department of Medical Oncology, Thomas Jefferson University Hospital, Philadelphia
| | - S Dubey
- Amgen Inc., South San Francisco
| | | | | | | | | | | | | | | | - S-P Choo
- Medical Oncology, National Cancer Centre Singapore, Singapore
| |
Collapse
|
45
|
Ibsen S, Su Y, Norton J, Zahavy E, Hayashi T, Adams S, Wrasidlo W, Esener S. Extraction protocol and mass spectrometry method for quantification of doxorubicin released locally from prodrugs in tumor tissue. JOURNAL OF MASS SPECTROMETRY : JMS 2013; 48:768-73. [PMID: 23832932 PMCID: PMC4110111 DOI: 10.1002/jms.3221] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 02/26/2013] [Accepted: 04/10/2013] [Indexed: 05/15/2023]
Abstract
The localized conversion of inactive doxorubicin prodrug chemotherapeutics to pharmacalogically active forms is difficult to quantify in mouse tumor models because it occurs only in small regions of tissue. The tumor tissue extraction protocol and LC-MS/MS analysis method described here were optimized to obtain a detection limit of 7.8 pg for the activated doxorubicin and 0.36 ng for the doxorubicin prodrug. This method can be useful for determining the biodistribution and activation efficiency for many different doxorubicin prodrugs. It can also be used for quantification of doxorubicin from tumor models that have poor vascularization resulting in low tissue accumulation.
Collapse
Affiliation(s)
- Stuart Ibsen
- Department of Bioengineering, Moores Cancer Center, University of California San Diego, 3855 Health Sciences Dr. # 0815, La Jolla, CA 92093-0815, USA.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Lim B, Scicchitano A, Beachler C, Gusani N, Sarwani N, Yang Z, Staveley-O'Carroll K, Ashkenazi A, Portera C, El-Deiry WS. FOLFIRI plus dulanermin (rhApo2L/TRAIL) in a patient with BRAF-mutant metastatic colon cancer. Cancer Biol Ther 2013; 14:711-9. [PMID: 23792567 DOI: 10.4161/cbt.25310] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Colorectal cancer patients with BRAF-mutant tumors have a more aggressive, rapidly progressing disease that is in critical need of novel therapeutic approaches. Indeed, whereas the median overall survival (OS) of colorectal cancer (CRC) patients receiving standard-of-care therapy is approximately two years or more if their tumors express wild-type BRAF and wild-type KRAS, median OS is less than twelve months with tumors expressing V600E-mutant BRAF and wild-type KRAS. Pro-apoptotic receptor agonists are a class of biologic agents under development to induce tumor-specific apoptosis and are being combined with classical chemotherapy or targeted agents in clinical trials. Herein, we present the case of a patient with bulky V600E-mutant BRAF hepatic flexure colon carcinoma, treated initially with FOLFOX plus bevacizumab neoadjuvant therapy and surgery. The patient had a rapid tumor relapse with metastatic disease to the liver and lung, and was enrolled in a phase 1b open-label clinical study, where he received the FOLFIRI regimen in combination with the pro-apoptotic receptor agonist dulanermin (rhApo2L/TRAIL). The patient maintained stable disease through 25 doses administered every two weeks before his disease progressed. After coming off study, the patient underwent surgical debulking and received intraperitoneal hyperthermic chemotherapy. He subsequently relapsed and was treated with FOLFIRI plus cetuximab. At the time of this report, the patient remains on active treatment. It is unclear what effect dulanermin may have had on the course of his disease, but it is noteworthy that the patient remained on FOLFIRI plus dulanermin therapy for a period that exceeded the median OS for patients with advanced, aggressive BRAF-mutant CRC. It is also noteworthy that at the time of this report the patient's overall survival since diagnosis has exceeded 30 months, which is beyond what is generally observed even for patients with CRC harboring wild-type BRAF and wild-type KRAS.
Collapse
Affiliation(s)
- Bora Lim
- Hematology/Oncology Division, Penn State Hershey Cancer Institute, Penn State Hershey Medical Center, Hershey, PA USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Li F, Zhao C, Wang L. Molecular-targeted agents combination therapy for cancer: developments and potentials. Int J Cancer 2013; 134:1257-69. [PMID: 23649791 DOI: 10.1002/ijc.28261] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 04/25/2013] [Indexed: 11/10/2022]
Abstract
Although chemotherapy has advanced into the era of targeted drugs, the antitumor efficacies of current therapies are limited, most likely because of the high degree of cancer clonal heterogeneity, intratumor genetic heterogeneity and cell signal complexity. As shutdown of a single target does not necessarily eradicate the cancer, the use of combinations of molecular-targeted agents (MATs) has been proposed, and some pioneering research has been conducted to examine the efficacy of this strategy. In this article, the clinical and preclinical studies that are underway in an attempt to improve the anticancer efficacy of chemotherapies through combination strategies are summarized. Studies of combining cytotoxic agents with MATs, coinhibiting two or more targets in a single pathway or coinhibiting parallel or compensatory pathways as well as specific combinations will be introduced, and the antitumor potentials of each combination strategy will be evaluated.
Collapse
Affiliation(s)
- Feifei Li
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, Beijing, China; Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing, China; Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, China
| | | | | |
Collapse
|
48
|
Pishas KI, Neuhaus SJ, Clayer MT, Adwal A, Brown MP, Evdokiou A, Callen DF, Neilsen PM. Pre-activation of the p53 pathway through Nutlin-3a sensitises sarcomas to drozitumab therapy. Oncol Rep 2013; 30:471-7. [PMID: 23670273 DOI: 10.3892/or.2013.2454] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 03/20/2013] [Indexed: 11/06/2022] Open
Abstract
The present study evaluated the efficacy of drozitumab, a human monoclonal agonistic antibody directed against death receptor 5 (DR5), as a new therapeutic avenue for the targeted treatment of bone and soft-tissue sarcomas. The antitumour activity of drozitumab as a monotherapy or in combination with Nutlin-3a was evaluated in a panel of sarcoma cell lines in vitro and human sarcoma patient samples ex vivo. Knockdown experiments were used to investigate the central role of p53 as a regulator of drozitumab cytotoxicity. Pre-activation of the p53 pathway through Nutlin-3a upregulated DR5, subsequently sensitising sarcoma cell lines and human sarcoma specimens to the pro-apoptotic effects of drozitumab. Silencing of p53 strongly decreased DR5 mRNA expression resulting in abrogation of drozitumab-induced apoptosis. Our study provides the first pre-clinical evaluation of combination therapy using p53-activating agents with drozitumab to further sensitise sarcomas to the cytotoxic effects of DR5 antibody therapy.
Collapse
Affiliation(s)
- Kathleen I Pishas
- Centre for Personalised Cancer Medicine, The University of Adelaide, Adelaide, South Australia, Australia
| | | | | | | | | | | | | | | |
Collapse
|
49
|
den Hollander MW, Gietema JA, de Jong S, Walenkamp AM, Reyners AK, Oldenhuis CN, de Vries EG. Translating TRAIL-receptor targeting agents to the clinic. Cancer Lett 2013; 332:194-201. [DOI: 10.1016/j.canlet.2012.04.007] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 04/11/2012] [Accepted: 04/15/2012] [Indexed: 12/21/2022]
|
50
|
Sharma S, de Vries EG, Infante JR, Oldenhuis CN, Gietema JA, Yang L, Bilic S, Parker K, Goldbrunner M, Scott JW, Burris HA. Safety, pharmacokinetics, and pharmacodynamics of the DR5 antibody LBY135 alone and in combination with capecitabine in patients with advanced solid tumors. Invest New Drugs 2013; 32:135-44. [PMID: 23589214 DOI: 10.1007/s10637-013-9952-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 03/15/2013] [Indexed: 12/01/2022]
Abstract
PURPOSE We evaluated the safety, maximum tolerated dose (MTD), pharmacokinetics, pharmacodynamics, biologic activity, and antitumor efficacy of the DR5 antibody, LBY135 ± capecitabine. EXPERIMENTAL DESIGN Escalating LBY135 was administered every 21 days, alone (Arm1) or with capecitabine (Arm2), to patients with advanced solid tumors. RESULTS In Arm1 (n = 40), LBY135 (0.3-40 mg/kg) resulted in no dose-limiting toxicities (DLTs); adverse events (AEs) included fatigue, hypotension, abdominal pain, dyspnea, and nausea. Stable disease (SD) was observed in 21/38 (55.3 %) patients. In Arm2 (n = 33), LBY135 (1-40 mg/kg) plus capecitabine resulted in 3 DLTs (each grade 3): dehydration and mucosal inflammation (1 mg/kg), colitis (20 mg/kg), and diarrhea (40 mg/kg). AEs included fatigue, nausea, dyspnea, and vomiting. Partial response was observed in 2 patients (rectal and breast cancer) and SD in 12/27 (44.4 %) patients. Mean elimination half-life of LBY135 ± capecitabine at saturation of clearance (≥10 mg/kg) ranged between 146 h and 492 h. Immunogenicity was detected in 16/73 (22 %) patients, of which 6 patients experienced reduced LBY135 exposure with repeat dosing. M30/M65 levels were not predictive for LBY135 response. FDG-PET responses were not consistently associated with RECIST responses. CONCLUSIONS LBY135 was well tolerated up to 40 mg/kg, the maximal dose administered; no MTD for LBY135 ± capecitabine was defined. Clearance was saturated at doses ≥10 mg/kg.
Collapse
Affiliation(s)
- Sunil Sharma
- Huntsman Cancer Institute, Division of Medical Oncology, University of Utah, 2000 Circle of Hope Drive, Suite 3380, Salt Lake City, UT, 84112, USA,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|