151
|
First-in-Human Phase I Study of Aprutumab Ixadotin, a Fibroblast Growth Factor Receptor 2 Antibody-Drug Conjugate (BAY 1187982) in Patients with Advanced Cancer. Target Oncol 2020; 14:591-601. [PMID: 31502117 PMCID: PMC6797631 DOI: 10.1007/s11523-019-00670-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Fibroblast growth factor receptor (FGFR) 2 is overexpressed in several tumor types, including triple-negative breast cancer and gastric cancer, both of which have a high unmet medical need. Aprutumab ixadotin (BAY 1187982) is the first antibody–drug conjugate (ADC) to target FGFR2 and the first to use a novel auristatin-based payload. Objective This first-in-human trial was conducted to determine the safety, tolerability, and maximum tolerated dose (MTD) of aprutumab ixadotin in patients with advanced solid tumors from cancer indications known to be FGFR2-positive. Patients and Methods In this open-label, multicenter, phase I dose-escalation trial (NCT02368951), patients with advanced solid tumors received escalating doses of aprutumab ixadotin (starting at 0.1 mg/kg body weight), administered intravenously on day 1 of every 21-day cycle. Primary endpoints included safety, tolerability, and the MTD of aprutumab ixadotin; secondary endpoints were pharmacokinetic evaluation and tumor response to aprutumab ixadotin. Results Twenty patients received aprutumab ixadotin across five cohorts, at doses of 0.1–1.3 mg/kg. The most common grade ≥ 3 drug-related adverse events were anemia, aspartate aminotransferase increase, proteinuria, and thrombocytopenia. Dose-limiting toxicities were thrombocytopenia, proteinuria, and corneal epithelial microcysts, and were only seen in the two highest dosing cohorts. The MTD was determined to be 0.2 mg/kg due to lack of quantitative data following discontinuations at 0.4 and 0.8 mg/kg doses. One patient had stable disease; no responses were reported. Conclusions Aprutumab ixadotin was poorly tolerated, with an MTD found to be below the therapeutic threshold estimated preclinically; therefore, the trial was terminated early. ClinicalTrials.gov Identifier NCT02368951. Electronic supplementary material The online version of this article (10.1007/s11523-019-00670-4) contains supplementary material, which is available to authorized users.
Collapse
|
152
|
Hassan R, Blumenschein GR, Moore KN, Santin AD, Kindler HL, Nemunaitis JJ, Seward SM, Thomas A, Kim SK, Rajagopalan P, Walter AO, Laurent D, Childs BH, Sarapa N, Elbi C, Bendell JC. First-in-Human, Multicenter, Phase I Dose-Escalation and Expansion Study of Anti-Mesothelin Antibody-Drug Conjugate Anetumab Ravtansine in Advanced or Metastatic Solid Tumors. J Clin Oncol 2020; 38:1824-1835. [PMID: 32213105 PMCID: PMC7255978 DOI: 10.1200/jco.19.02085] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE This phase I study, which to our knowledge is the first-in-human study of this kind, investigates the safety, tolerability, pharmacokinetics, and clinical activity of anetumab ravtansine, an antibody–drug conjugate of anti-mesothelin antibody linked to maytansinoid DM4, in patients with advanced, metastatic, or recurrent solid tumors known to express the tumor-differentiation antigen mesothelin. PATIENTS AND METHODS This phase I, open-label, multicenter, dose-escalation and dose-expansion study of anetumab ravtansine enrolled 148 adult patients with multiple solid tumor types. Ten dose-escalation cohorts of patients with advanced or metastatic solid tumors (0.15-7.5 mg/kg) received anetumab ravtansine once every 3 weeks, and 6 expansion cohorts of patients with advanced, recurrent ovarian cancer or malignant mesothelioma received anetumab ravtansine at the maximum tolerated dose once every 3 weeks, 1.8 mg/kg once per week, and 2.2 mg/kg once per week. RESULTS Forty-five patients were enrolled across the 10 dose-escalation cohorts. The maximum tolerated dose of anetumab ravtansine was 6.5 mg/kg once every 3 weeks or 2.2 mg/kg once per week. Thirty-two patients were enrolled in the 6.5 mg/kg once-every-3-weeks, 35 in the 1.8 mg/kg once-per-week, and 36 in the 2.2 mg/kg once-per-week expansion cohorts. The most common drug-related adverse events were fatigue, nausea, diarrhea, anorexia, vomiting, peripheral sensory neuropathy, and keratitis/keratopathy. There were no drug-related deaths. Anetumab ravtansine pharmacokinetics were dose proportional; the average half-life was 5.5 days. Among 148 patients with mesothelioma or ovarian, pancreatic, non–small-cell lung, and breast cancers, 1 had a complete response, 11 had partial responses, and 66 had stable disease. High levels of tumor mesothelin expression were detected in patients with clinical activity. CONCLUSION Anetumab ravtansine exhibited a manageable safety and favorable pharmacokinetic profile with encouraging preliminary antitumor activity in heavily pretreated patients with mesothelin-expressing solid tumors. The results allowed for the determination of recommended doses, schedules, and patient populations for anetumab ravtansine in phase II studies.
Collapse
Affiliation(s)
- Raffit Hassan
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - George R Blumenschein
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kathleen N Moore
- Stephenson Oklahoma Cancer Center at University of Oklahoma, Oklahoma City, OK/Sarah Cannon Research Institute, Nashville, TN
| | | | | | - John J Nemunaitis
- Division of Hematology and Medical Oncology, Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Shelly M Seward
- Wayne State University Karmanos Cancer Institute, Huntington Woods, MI
| | - Anish Thomas
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | | | | | | | | | | | | | - Cem Elbi
- Bayer HealthCare Pharmaceuticals, Whippany, NJ
| | | |
Collapse
|
153
|
Popat R, Warcel D, O'Nions J, Cowley A, Smith S, Tucker WR, Yong K, Esposti SD. Characterization of response and corneal events with extended follow-up after belantamab mafodotin (GSK2857916) monotherapy for patients with relapsed multiple myeloma: a case series from the first-time-in-human clinical trial. Haematologica 2020; 105:e261-e263. [PMID: 32107339 DOI: 10.3324/haematol.2019.235937] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Rakesh Popat
- National Institute for Health Research Clinical Research Facility, University College London Hospitals NHS Foundation Trust
| | - Dana Warcel
- National Institute for Health Research Clinical Research Facility, University College London Hospitals NHS Foundation Trust
| | - Jenny O'Nions
- National Institute for Health Research Clinical Research Facility, University College London Hospitals NHS Foundation Trust
| | - Anna Cowley
- National Institute for Health Research Clinical Research Facility, University College London Hospitals NHS Foundation Trust
| | - Sasha Smith
- National Institute for Health Research Clinical Research Facility, University College London Hospitals NHS Foundation Trust
| | - William R Tucker
- National Institute for Health Research Biomedical Research Centre for Ophthalmology at Moorfields Eye Hospital NHS Foundation Trust & University College London Institute of Ophthalmology, London, UK
| | - Kwee Yong
- National Institute for Health Research Clinical Research Facility, University College London Hospitals NHS Foundation Trust
| | - Simona Degli Esposti
- National Institute for Health Research Biomedical Research Centre for Ophthalmology at Moorfields Eye Hospital NHS Foundation Trust & University College London Institute of Ophthalmology, London, UK
| |
Collapse
|
154
|
Birrer MJ, Moore KN, Betella I, Bates RC. Antibody-Drug Conjugate-Based Therapeutics: State of the Science. J Natl Cancer Inst 2020; 111:538-549. [PMID: 30859213 DOI: 10.1093/jnci/djz035] [Citation(s) in RCA: 278] [Impact Index Per Article: 55.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/21/2019] [Accepted: 03/08/2019] [Indexed: 02/06/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are complex engineered therapeutics consisting of monoclonal antibodies, directed toward tumor-associated antigens, to which highly potent cytotoxic agents are attached using chemical linkers. This targeted drug delivery strategy couples the precision of the antibody targeting moiety with the cytocidal activity of the payload, which is generally too toxic on its own to be systemically administered. In this manner, ADCs confer a means to reduce off-target toxicities in patients by limiting the exposure of normal tissues to the payload, thus broadening the potential therapeutic window compared with traditional chemotherapy. The pace of ADC development is accelerating, with the number of investigational agents in human trials having more than tripled over the past 5 years, underscoring the enthusiasm for this transformative approach to cancer treatment. Here, we review the key structural elements of ADC design (antibody, linker, and payload), highlighting critical aspects and technological advances that have affected the clinical effectiveness of this class of biopharmaceuticals. The ADC field continues to evolve, including ongoing efforts aimed at improving target selection, developing payloads with varied mechanisms of action and increased potency, designing innovative bioconjugation strategies, as well as maximizing efficacy and tolerability in patients. An overview of the current clinical trial landscape is provided, with emphasis on the clinical experience of the four ADCs to have received regulatory approval to date, as well as additional promising candidates currently in late-stage clinical development in both solid tumor and hematological malignancies.
Collapse
Affiliation(s)
- Michael J Birrer
- Division of Hematology-Oncology, University of Alabama at Birmingham Comprehensive Cancer Center, Birmingham, AL
| | - Kathleen N Moore
- Stephenson Department of Obstetrics and Gynecology, Oklahoma Cancer Center at the University of Oklahoma Health Sciences Center, Oklahoma City, OK.,Sarah Cannon Research Institute, Nashville, TN
| | - Ilaria Betella
- Division of Hematology-Oncology, University of Alabama at Birmingham Comprehensive Cancer Center, Birmingham, AL
| | | |
Collapse
|
155
|
Lonial S, Lee HC, Badros A, Trudel S, Nooka AK, Chari A, Abdallah AO, Callander N, Lendvai N, Sborov D, Suvannasankha A, Weisel K, Karlin L, Libby E, Arnulf B, Facon T, Hulin C, Kortüm KM, Rodríguez-Otero P, Usmani SZ, Hari P, Baz R, Quach H, Moreau P, Voorhees PM, Gupta I, Hoos A, Zhi E, Baron J, Piontek T, Lewis E, Jewell RC, Dettman EJ, Popat R, Esposti SD, Opalinska J, Richardson P, Cohen AD. Belantamab mafodotin for relapsed or refractory multiple myeloma (DREAMM-2): a two-arm, randomised, open-label, phase 2 study. Lancet Oncol 2020; 21:207-221. [DOI: 10.1016/s1470-2045(19)30788-0] [Citation(s) in RCA: 359] [Impact Index Per Article: 71.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/18/2019] [Accepted: 11/26/2019] [Indexed: 12/17/2022]
|
156
|
Shin E, Lim DH, Han J, Nam DH, Park K, Ahn MJ, Kang WK, Lee J, Ahn JS, Lee SH, Sun JM, Jung HA, Chung TY. Markedly increased ocular side effect causing severe vision deterioration after chemotherapy using new or investigational epidermal or fibroblast growth factor receptor inhibitors. BMC Ophthalmol 2020; 20:19. [PMID: 31918686 PMCID: PMC6953164 DOI: 10.1186/s12886-019-1285-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 12/23/2019] [Indexed: 12/25/2022] Open
Abstract
Background We sought to describe corneal epithelial changes after using epidermal (EGFR) or fibroblast growth factor receptor (FGFR) inhibitors as chemotherapy and to clarify incidence and prognosis. Materials Retrospective chart review. Results Among 6871 patients and 17 EGFR or FGFR inhibitors, 1161 patients (16.9%) referred for ophthalmologic examination. In total, 1145 patients had disease-related or unrelated ocular complications. Among 16 patients with treatment-related ocular complications, three patients had treatment-related radiation retinopathy and one patient showed treatment-related corneal ulcer. Finally the authors identified that, in 12 patients, three EGFR inhibitors and two FGFR inhibitors caused corneal epithelial lesions. Vandetanib, Osimertinib, and ABT-414 caused vortex keratopathy in nine patients, while ASP-5878 and FPA-144 caused epithelial changes resembling corneal dysmaturation in three patients. The mean interval until symptoms appeared was 246 days with vandetanib, 196 days with osimertinib, 30 days with ABT-414, 55 days with ASP-5878, and 70 days with FPA-144. The mean of the lowest logarithm of minimal angle of resolution visual acuity results of the right and left eyes after chemotherapy were 0.338 and 0.413. The incidence rates of epithelial changes were 15.79% with vandetanib, 0.5% with osimertinib, 100% with ABT-414, 50.0% with ASP-5878, and 18.2% with FPA-144. After excluding deceased patients and those who were lost to follow-up or still undergoing treatment, we confirmed the reversibility of corneal lesions after the discontinuation of each agent. Seven patients showed full recovery of their vision and corneal epithelium, while three achieved a partial level of recovery. Although patients diagnosed with glioblastoma used prophylactic topical steroids before and during ABT-414 therapy, all developed vortex keratopathy. Conclusions EGFR and FGFR inhibitors are chemotherapy agents that could make corneal epithelial changes. Contrary to the low probability of ocular complication with old EGFR drugs, recently introduced EGFR and FGFR agents showed a high incidence of ocular complication with severe vision distortion. Doctors should forewarn patients planning chemotherapy with these agents that decreased visual acuity could develop due to corneal epithelial changes and also reassure them that the condition could be improved after the end of treatment without the use of steroid eye drops. Trial registration This study was approved by the institutional review board (IRB) of Samsung Medical Center (IRB no. 2019–04-027) and was conducted according to the principles expressed in the Declaration of Helsinki.
Collapse
Affiliation(s)
- Eunhae Shin
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Dong Hui Lim
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea. .,Department of Preventive Medicine, Graduate School, Catholic University of Korea, Seoul, Republic of Korea.
| | - Jisang Han
- Department of Ophthalmology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Do-Hyun Nam
- Cancer Stem Cell Research Center, Department of Neurosurgery, Samsung Medical Center and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Keunchil Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Won Ki Kang
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jeeyun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jong-Mu Sun
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyun Ae Jung
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Tae-Young Chung
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
157
|
Abstract
PURPOSE OF REVIEW Antibody drug conjugates (ADC) are a novel class of cancer therapeutics, delivering cytotoxic therapy directly to cancer cells, and show promise in the management of platinum-resistant ovarian cancer. Herein we summarize the ADC landscape currently in clinical study. RECENT FINDINGS Mirvetuximab Soravtansine, IMGN853, is an ADC targeting the folate receptor alpha (FRα) and has demonstrated promising single agent activity and a favorable toxicity profile in FRα-positive, platinum-resistant, epithelial ovarian cancer (EOC). The antitumor effect is seen primarily in less heavily pretreated EOC patients with moderate-to-high FRα tumor expression. A phase III study, randomizing patients to either IMGN853 or the physician's choice of single-agent chemotherapy has completed accrual. Additional ADC are being evaluated in ovarian cancer including agents that target NaPiB2, Trop2, mesothelin, and MUC16 are in phase 1 clinical trials. SUMMARY ADC bind antigens overexpressed on cancer cells and provide site-selective drug delivery, with the goal to increase therapeutic efficacy of cytotoxics while decreasing the off-target toxicity of the payloads. With appropriate antigen selection and adequate, measurable antigen threshold targets, these new agents may provide an improved strategy for overcoming resistance to standard chemotherapy in ovarian cancer.
Collapse
|
158
|
Mahalingaiah PK, Ciurlionis R, Durbin KR, Yeager RL, Philip BK, Bawa B, Mantena SR, Enright BP, Liguori MJ, Van Vleet TR. Potential mechanisms of target-independent uptake and toxicity of antibody-drug conjugates. Pharmacol Ther 2019; 200:110-125. [DOI: 10.1016/j.pharmthera.2019.04.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/19/2019] [Indexed: 12/13/2022]
|
159
|
Banerji U, van Herpen CML, Saura C, Thistlethwaite F, Lord S, Moreno V, Macpherson IR, Boni V, Rolfo C, de Vries EGE, Rottey S, Geenen J, Eskens F, Gil-Martin M, Mommers EC, Koper NP, Aftimos P. Trastuzumab duocarmazine in locally advanced and metastatic solid tumours and HER2-expressing breast cancer: a phase 1 dose-escalation and dose-expansion study. Lancet Oncol 2019; 20:1124-1135. [PMID: 31257177 DOI: 10.1016/s1470-2045(19)30328-6] [Citation(s) in RCA: 384] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/23/2019] [Accepted: 04/26/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Trastuzumab duocarmazine is a novel HER2-targeting antibody-drug conjugate comprised of trastuzumab covalently bound to a linker drug containing duocarmycin. Preclinical studies showed promising antitumour activity in various models. In this first-in-human study, we assessed the safety and activity of trastuzumab duocarmazine in patients with advanced solid tumours. METHODS We did a phase 1 dose-escalation and dose-expansion study. The dose-escalation cohort comprised patients aged 18 years or older enrolled from three academic hospitals in Belgium, the Netherlands, and the UK with locally advanced or metastatic solid tumours with variable HER2 status who were refractory to standard cancer treatment. A separate cohort of patients were enrolled to the dose-expansion phase from 15 hospitals in Belgium, the Netherlands, Spain, and the UK. Dose-expansion cohorts included patients aged 18 years or older with breast, gastric, urothelial, or endometrial cancer with at least HER2 immunohistochemistry 1+ expression and measurable disease according to Response Evaluation Criteria in Solid Tumors (RECIST). Trastuzumab duocarmazine was administered intravenously on day 1 of each 3-week cycle. In the dose-escalation phase, trastuzumab duocarmazine was given at doses of 0·3 mg/kg to 2·4 mg/kg (3 + 3 design) until disease progression or unacceptable toxicity. The primary endpoint of the dose-escalation phase was to assess safety and ascertain the recommended phase 2 dose, which would be the dose used in the dose-expansion phase. The primary endpoint of the dose-expansion phase was the proportion of patients achieving an objective response (complete response or partial response), as assessed by the investigator using RECIST version 1.1. This ongoing study is registered with ClinicalTrials.gov, number NCT02277717, and is fully recruited. FINDINGS Between Oct 30, 2014, and April 2, 2018, 39 patients were enrolled and treated in the dose-escalation phase and 146 patients were enrolled and treated in the dose-expansion phase. One dose-limiting toxic effect (death from pneumonitis) occurred at the highest administered dose (2·4 mg/kg) in the dose-escalation phase. One further death occurred in the dose-escalation phase (1·5 mg/kg cohort) due to disease progression, which was attributed to general physical health decline. Grade 3-4 treatment-related adverse events reported more than once in the dose-escalation phase were keratitis (n=3) and fatigue (n=2). Based on all available data, the recommended phase 2 dose was set at 1·2 mg/kg. In the dose-expansion phase, treatment-related serious adverse events were reported in 16 (11%) of 146 patients, most commonly infusion-related reactions (two [1%]) and dyspnoea (two [1%]). The most common treatment-related adverse events (grades 1-4) were fatigue (48 [33%] of 146 patients), conjunctivitis (45 [31%]), and dry eye (45 [31%]). Most patients (104 [71%] of 146) had at least one ocular adverse event, with grade 3 events reported in ten (7%) of 146 patients. No patients died from treatment-related adverse events and four patients died due to disease progression, which were attributed to hepatic failure (n=1), upper gastrointestinal haemorrhage (n=1), neurological decompensation (n=1), and renal failure (n=1). In the breast cancer dose-expansion cohorts, 16 (33%, 95% CI 20·4-48·4) of 48 assessable patients with HER2-positive breast cancer achieved an objective response (all partial responses) according to RECIST. Nine (28%, 95% CI 13·8-46·8) of 32 patients with HER2-low, hormone receptor-positive breast cancer and six (40%, 16·3-67·6) of 15 patients with HER2-low, hormone receptor-negative breast cancer achieved an objective response (all partial responses). Partial responses were also observed in one (6%, 95% CI 0·2-30·2) of 16 patients with gastric cancer, four (25%, 7·3-52·4) of 16 patients with urothelial cancer, and five (39%, 13·9-68·4) of 13 patients with endometrial cancer. INTERPRETATION Trastuzumab duocarmazine shows notable clinical activity in heavily pretreated patients with HER2-expressing metastatic cancer, including HER2-positive trastuzumab emtansine-resistant and HER2-low breast cancer, with a manageable safety profile. Further investigation of trastuzumab duocarmazine for HER2-positive breast cancer is ongoing and trials for HER2-low breast cancer and other HER2-expressing cancers are in preparation. FUNDING Synthon Biopharmaceuticals.
Collapse
Affiliation(s)
- Udai Banerji
- Institute of Cancer Research and The Royal Marsden, London, UK.
| | | | - Cristina Saura
- Vall d'Hebrón University Hospital, Vall d'Hebrón Institute of Oncology, Barcelona, Spain
| | - Fiona Thistlethwaite
- The Christie NHS Foundation Trust and The University of Manchester, Manchester, UK
| | | | | | | | | | - Christian Rolfo
- University Hospital Antwerp, Edegem, Belgium; Greenebaum Comprehensive Cancer Center, Maryland University, Baltimore, MD, USA
| | | | | | - Jill Geenen
- The Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | | | | | | | - Philippe Aftimos
- Institut Jules Bordet-Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
160
|
Thackaberry EA, Lorget F, Farman C, Bantseev V. The safety evaluation of long-acting ocular delivery systems. Drug Discov Today 2019; 24:1539-1550. [DOI: 10.1016/j.drudis.2019.05.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/09/2019] [Accepted: 05/31/2019] [Indexed: 01/14/2023]
|
161
|
Tai YT, Anderson KC. B cell maturation antigen (BCMA)-based immunotherapy for multiple myeloma. Expert Opin Biol Ther 2019; 19:1143-1156. [PMID: 31277554 DOI: 10.1080/14712598.2019.1641196] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: B cell maturation antigen (BCMA) contributes to MM pathophysiology and is a target antigen for novel MM immunotherapy. Complete responses have been observed in heavily pretreated MM patients after treatment with BCMA antibody-drug conjugates (ADC), chimeric antigen receptor T, and bi-specific T cell engagers (BiTE®). These and other innovative BCMA-targeted therapies transform the treatment landscape and patient outcome in MM. Areas covered: The immunobiological rationale for targeting BCMA in MM is followed by key preclinical studies and available clinical data on efficacy and safety of therapies targeting BCMA from recent phase I/II studies. Expert opinion: BCMA is the most selective MM target antigen, and BCMA-targeted approaches have achieved high responses even in relapse and refractory MM as a monotherapy. Long-term follow-up and correlative studies using immuno-phenotyping and -sequencing will delineate mechanisms of overcoming the immunosuppressive MM bone marrow microenvironment to mediate additive or synergistic anti-MM cytotoxicity. Moreover, they will delineate cellular and molecular events underlying the development of resistance underlying relapse of disease. Most importantly, targeted BCMA-based immunotherapies used earlier in the disease course and in combination (adoptive T cell therapy, mAbs/ADCs, checkpoint and cytokine blockade, and vaccines) have great promise to achieve long-term disease control and potential cure.
Collapse
Affiliation(s)
- Yu-Tzu Tai
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School , Boston , MA , USA
| | - Kenneth C Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School , Boston , MA , USA
| |
Collapse
|
162
|
Ma H, Sawas A. Combining Biology and Chemistry for a New Take on Chemotherapy: Antibody-Drug Conjugates in Hematologic Malignancies. Curr Hematol Malig Rep 2019; 13:555-569. [PMID: 30362019 DOI: 10.1007/s11899-018-0485-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE OF REVIEW This review is about the antibody-drug conjugate (ADC), a form of drug delivery consisting of a monoclonal antibody, linker, and cytotoxic payload. We summarize the history of ADC development, highlighting the three FDA-approved ADCs currently available. RECENT FINDINGS Gemtuzumab ozogamicin is a CD33-targeted ADC linked to calicheamicin. It is approved for CD33+ AML in the first line or the relapsed or refractory (R/R) setting. Brentuximab vedotin is a CD30-targeted ADC bound to MMAE. It is approved for the treatment of certain R/R CD30+ lymphomas. Recently, it has been approved for first line therapy with chemotherapy in advanced HL. Inotuzumab ozogamicin is a CD22-directed ADC attached to calicheamicin indicated for the treatment of adults with R/R B cell precursor ALL. Three ADCs have been approved for the treatment of various hematologic malignancies. We discuss the pertinent human trials that led to FDA approval. We include our perspectives about drug resistance, toxicities, and future development.
Collapse
Affiliation(s)
- Helen Ma
- Columbia University Medical Center, New York, NY, 10032, USA
| | - Ahmed Sawas
- Columbia University Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
163
|
Kunkler AL, Binkley EM, Mantopoulos D, Hendershot AJ, Ohr MP, Kendra KL, Davidorf FH, Cebulla CM. Known and novel ocular toxicities of biologics, targeted agents, and traditional chemotherapeutics. Graefes Arch Clin Exp Ophthalmol 2019; 257:1771-1781. [PMID: 31098752 DOI: 10.1007/s00417-019-04337-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 02/14/2019] [Accepted: 04/22/2019] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Increases in cancer with an aging population and the rapid development of new chemotherapeutics underscore the need for ophthalmologists to identify and manage potential ocular toxicities. This retrospective case series reports the ocular side effects of traditional and novel chemotherapeutic agents from a large center. METHODS The medical records of 3537 adult patients 18 years and older who presented to an academic ophthalmology department on high-risk medications identified by ICD-9 search between January 2010 and February 2015 were reviewed. A cancer diagnosis, as well as a temporal association with chemotherapeutic use and ocular side effect, was deemed necessary for inclusion in the study. The main measures were ocular side effects in cancer patients taking chemotherapy, ocular imaging abnormalities, and the outcome of each side effect. RESULTS Of the 161 oncology patients referred to the ophthalmology clinic for chemotherapeutic screening or ocular side effect, 31 (19.3%) were identified as having an ocular adverse reaction due to a novel or traditional chemotherapeutic medication. A novel flattening of the corneal curvature with hyperopic shift and corneal microcysts was identified in a patient taking the antibody-drug conjugate mirvetuximab soravtansine and was reversible with topical steroids. A bilateral medium-vessel choroidal vasculopathy with serous retinal detachment was seen with ipilimumab. The most frequent medication with ocular toxicity was interferon-α(2b) (IFN-α(2b)) (6/31, 19.4%); headache was typical in these patients (83.3%). Ibrutinib ocular toxicity was second most common (5/31, 16.1%), usually causing red or dry eye, while one patient developed branch retinal artery occlusion. Retinal abnormalities documented on OCT imaging occurred with IFN-α(2b), ipilimumab, binimetinib, and docetaxel, while rod-cone ERG abnormality was seen with cisplatin. Inflammatory conditions included anterior scleritis with zoledronic acid, focal eyelid inflammation with veliparib, bilateral chemosis with R-CHOP, iritis, and blepharospasm with IFN-α(2b). AION occurred with pemetrexed, and transient vision loss with hyperemic disc OS was seen with FOLFOX. Two patients (2/31, 6.5%) developed permanent vision loss. Six patients were lost to follow-up, and the clinical course was unknown (6/31, 19.4%). CONCLUSIONS AND RELEVANCE Cases of permanent visual loss were observed; yet, in the majority of side effects, they improved with topical therapy and/or holding the medication. Further research is needed to elucidate the incidence and the pathophysiology of these side effects and maximize patient quality of life.
Collapse
Affiliation(s)
- Anne L Kunkler
- Department of Ophthalmology and Visual Science, Havener Eye Institute, The Ohio State University, Wexner Medical Center, 915 Olentangy River Rd, Ste 5000, Columbus, OH, 43212, USA
| | - Elaine M Binkley
- Department of Ophthalmology and Visual Science, Havener Eye Institute, The Ohio State University, Wexner Medical Center, 915 Olentangy River Rd, Ste 5000, Columbus, OH, 43212, USA
| | - Dimosthenis Mantopoulos
- Department of Ophthalmology and Visual Science, Havener Eye Institute, The Ohio State University, Wexner Medical Center, 915 Olentangy River Rd, Ste 5000, Columbus, OH, 43212, USA
| | - Andrew J Hendershot
- Department of Ophthalmology and Visual Science, Havener Eye Institute, The Ohio State University, Wexner Medical Center, 915 Olentangy River Rd, Ste 5000, Columbus, OH, 43212, USA
| | - Matthew P Ohr
- Department of Ophthalmology and Visual Science, Havener Eye Institute, The Ohio State University, Wexner Medical Center, 915 Olentangy River Rd, Ste 5000, Columbus, OH, 43212, USA
| | - Kari L Kendra
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Wexner Medical Center, Columbus, OH, USA
| | - Frederick H Davidorf
- Department of Ophthalmology and Visual Science, Havener Eye Institute, The Ohio State University, Wexner Medical Center, 915 Olentangy River Rd, Ste 5000, Columbus, OH, 43212, USA
| | - Colleen M Cebulla
- Department of Ophthalmology and Visual Science, Havener Eye Institute, The Ohio State University, Wexner Medical Center, 915 Olentangy River Rd, Ste 5000, Columbus, OH, 43212, USA.
| |
Collapse
|
164
|
Li Z, Wang M, Yu D, Luo W, Fang J, Huang C, Yao X. Monomethyl auristatin E-conjugated anti-EGFR antibody inhibits the growth of human EGFR-positive non-small cell lung cancer. Cancer Chemother Pharmacol 2019; 84:61-72. [PMID: 31037333 DOI: 10.1007/s00280-019-03848-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 04/22/2019] [Indexed: 12/24/2022]
Abstract
PURPOSE Epidermal growth factor receptor (EGFR) is highly expressed on non-small cell lung cancers (NSCLC) and a valuable therapeutic target. This study aimed at producing and characterizing monomethyl auristatin E (MMAE)-conjugated anti-EGFR antibody as a novel EGFR-targeting therapy for NSCLC. METHODS A humanized anti-EGFR monoclonal antibody (named RC68) was purified and conjugated with MMAE using a MC-VC-PAB or PY-VC-PAB linker. The in vitro and in vivo antitumor activity of RC68-MC-VC-PAB-MMAE and RC68-PY-VC-PAB-MMAE were characterized. RESULTS The RC68 was generated from RC68-expressing cells and had a purity of > 99.0%. The RC68 recognized EGFR on tumor cells, particularly for higher EGFR expressing H125, A431, HCC827 and H1975 cells. The RC68 was conjugated with an average of 4 MMAE molecules to generate RC68-MC-VC-PAB-MMAE and RC68-PY-VC-PAB-MMAE, respectively. The RC68-MC-VC-PAB-MMAE, RC68-PY-VC-PAB-MMAE and RC68 displayed similar binding affinity to EGFR on tumor cells, and RC68-MC-VC-PAB-MMAE and RC68-PY-VC-PAB-MMAE were effectively internalized by H125 cells. The RC68-MC-VC-PAB-MMAE and RC68-PY-VC-PAB-MMAE inhibited the growth of H125 cells in vitro with an IC50 7.37-8.04 ng/mL and implanted H125 tumors in vivo, but did not affect body weights of mice. The antitumor effect of RC68-MC-VC-PAB-MMAE was stronger than RC68-PY-VC-PAB-MMAE, which was also stronger than docetaxel in vivo. CONCLUSIONS These novel antibody-drug conjugates, particularly for RC68-MC-VC-PAB-MMAE, may be a potential candidate for treatment of EGFR + NSCLC.
Collapse
Affiliation(s)
| | - Mingxue Wang
- Mabplex International Ltd., Yantai, 264006, Shandong, China
| | - Deling Yu
- Mabplex International Ltd., Yantai, 264006, Shandong, China
| | - Wenting Luo
- RemeGen, Ltd., Yantai, 264006, Shandong, China
| | - Jianmin Fang
- RemeGen, Ltd., Yantai, 264006, Shandong, China
- Mabplex International Ltd., Yantai, 264006, Shandong, China
- School of Life Science and Technology, Tongji University, Shanghai, 200092, China
| | | | - Xuejing Yao
- RemeGen, Ltd., Yantai, 264006, Shandong, China.
| |
Collapse
|
165
|
Antibody-drug conjugate, GSK2857916, in relapsed/refractory multiple myeloma: an update on safety and efficacy from dose expansion phase I study. Blood Cancer J 2019; 9:37. [PMID: 30894515 PMCID: PMC6426965 DOI: 10.1038/s41408-019-0196-6] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/15/2019] [Accepted: 02/25/2019] [Indexed: 12/24/2022] Open
Abstract
Interim analyses of a phase I study with GSK2857916, an antibody-drug conjugate against B cell maturation antigen, have previously reported a 60% overall response and 7.9 months progression-free survival in relapsed/refractory multiple myeloma (MM). We provide updated safety and efficacy results of the BMA117159 trial following an additional 14 months' follow-up. This open-label, first-in-human, phase I study was conducted at nine centres in the USA, Canada and the UK, and included adults with MM and progressive disease after stem cell transplantation, alkylators, proteasome inhibitors, and immunomodulators. In part 1, the recommended dose of 3.4 mg/kg was identified; in part 2, patients received GSK2857916 3.4 mg/kg once every 3 weeks. Selected part 2 safety/tolerability and efficacy endpoints are reported. Twenty-one (60.0%; 95% confidence interval (CI) 42.1-76.1) of 35 patients achieved partial response or better, including two stringent complete responses and three complete responses. The median progression-free survival was 12 months and median duration of response was 14.3 months. Thrombocytopenia and corneal events were commonly reported; no new safety signals were identified. GSK2857916 was well tolerated and demonstrated a rapid, deep and durable response in heavily pre-treated patients with relapsed/refractory MM, consolidating the interim analyses conclusions that GSK2857916 is a promising treatment for these patients.
Collapse
|
166
|
Deklerck E, Denys H, Kreps EO. Corneal features in trastuzumab emtansine treatment: not a rare occurrence. Breast Cancer Res Treat 2019; 175:525-530. [PMID: 30820718 DOI: 10.1007/s10549-019-05179-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 02/19/2019] [Indexed: 10/27/2022]
Abstract
PURPOSE Ado-trastuzumab emtansine (T-DM1/Kadcyla®;Genentech) is an antibody-drug conjugate used in the treatment of human epidermal growth factor receptor-2-positive metastasized breast cancer. Few studies report a spectrum of corneal changes in patients treated with this drug. Our aim is to specify the nature and prevalence of corneal features of T-DM1 treatment in order to formulate guidelines as to which findings necessitate systemic treatment cessation or dose reduction. METHODS We performed a cross-sectional, prospective study in all patients currently treated with T-DM1 or recently stopped in Ghent University Hospital, Belgium. RESULTS A total of 12 patients completed a full ophthalmic workup. Ten patients were currently using T-DM1, and two patients had recently (< 10 weeks) stopped treatment because of clinical non-response. Twenty eyes of 10 patients currently on T-DM1-treatment all exhibited coarse cystoid lesions to the deep corneal epithelial cells, primarily in the midperipheral area, both biomicroscopically and on confocal microscopy. The two patients who stopped treatment, displayed no corneal epithelial changes. Only three patients reported symptoms which were attributed to other ocular factors, likely not to be related to T-DM1 treatment. CONCLUSIONS This case series shows that asymptomatic, low-grade corneal epithelial changes are hallmark features in T-DM1-treatment and should not alarm clinicians. These findings are relatively stationary, reversible and thus do not require ocular treatment or cessation of systemic treatment.
Collapse
Affiliation(s)
- Els Deklerck
- Department of Ophthalmology, Ghent University Hospital, 10, Corneel Heymanslaan, 9000, Ghent, Belgium.
| | - Hannelore Denys
- Department of Medical Oncology, Ghent University Hospital, 10, Corneel Heymanslaan, 9000, Ghent, Belgium
| | - Elke O Kreps
- Department of Ophthalmology, Ghent University Hospital, 10, Corneel Heymanslaan, 9000, Ghent, Belgium
| |
Collapse
|
167
|
Lucas AT, Robinson R, Schorzman AN, Piscitelli JA, Razo JF, Zamboni WC. Pharmacologic Considerations in the Disposition of Antibodies and Antibody-Drug Conjugates in Preclinical Models and in Patients. Antibodies (Basel) 2019; 8:E3. [PMID: 31544809 PMCID: PMC6640706 DOI: 10.3390/antib8010003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/21/2018] [Accepted: 12/22/2018] [Indexed: 12/11/2022] Open
Abstract
The rapid advancement in the development of therapeutic proteins, including monoclonal antibodies (mAbs) and antibody-drug conjugates (ADCs), has created a novel mechanism to selectively deliver highly potent cytotoxic agents in the treatment of cancer. These agents provide numerous benefits compared to traditional small molecule drugs, though their clinical use still requires optimization. The pharmacology of mAbs/ADCs is complex and because ADCs are comprised of multiple components, individual agent characteristics and patient variables can affect their disposition. To further improve the clinical use and rational development of these agents, it is imperative to comprehend the complex mechanisms employed by antibody-based agents in traversing numerous biological barriers and how agent/patient factors affect tumor delivery, toxicities, efficacy, and ultimately, biodistribution. This review provides an updated summary of factors known to affect the disposition of mAbs/ADCs in development and in clinical use, as well as how these factors should be considered in the selection and design of preclinical studies of ADC agents in development.
Collapse
Affiliation(s)
- Andrew T Lucas
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Ryan Robinson
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Allison N Schorzman
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Joseph A Piscitelli
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | - Juan F Razo
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | - William C Zamboni
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
168
|
Xie H, Adjei AA. Antibody-Drug Conjugates for the Therapy of Thoracic Malignancies. J Thorac Oncol 2018; 14:358-376. [PMID: 30599202 DOI: 10.1016/j.jtho.2018.11.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/25/2018] [Accepted: 11/29/2018] [Indexed: 10/27/2022]
Abstract
Antibody-drug conjugates (ADCs) are a novel class of therapeutic agents incorporating both target-specific monoclonal antibodies and cytotoxic small molecules via a chemical linker. They were first introduced into the clinic for the treatment of advanced hematologic malignancies. The only approved ADC for solid tumors targets erb-b2 receptor tyrosine kinase (HER2), a validated antigen in breast cancer. Many ADCs are under active investigation for various types of solid tumors. In this article, we review the literature from several perspectives including the design, pharmacology, and mechanism-based toxicities of antibody-drug conjugates. We then discuss ADCs currently in clinical development for thoracic malignancies.
Collapse
Affiliation(s)
- Hao Xie
- Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
169
|
Kaye R, Botten N, Lippestad M, Li D, Hodges RR, Utheim TP, Serhan CN, Dartt DA. Resolvin D1, but not resolvin E1, transactivates the epidermal growth factor receptor to increase intracellular calcium and glycoconjugate secretion in rat and human conjunctival goblet cells. Exp Eye Res 2018; 180:53-62. [PMID: 30513286 DOI: 10.1016/j.exer.2018.11.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 11/16/2018] [Indexed: 12/19/2022]
Abstract
PURPOSE To identify interactions of the epidermal growth factor receptor (EGFR) with the pro-resolving mediator receptors for RvD1 and RvE1 to stimulate an increase in intracellular [Ca2+] ([Ca2+]i) and mucin secretion from cultured human and rat conjunctival goblet cells. METHODS Goblet cells from human and rat conjunctiva were grown in culture using RPMI media. Cultured goblet cells were pre-incubated with inhibitors, and then stimulated with RvD1, RvE1, EGF or the cholinergic agonist carbachol (Cch). Increase in [Ca2+]i was measured using fura-2/AM. Goblet cell secretion was measured using an enzyme-linked lectin assay with UEA-1. Western blot analysis was performed with antibodies against AKT and ERK 1/2. RESULTS In cultured human conjunctival goblet cells RvE1 -stimulated an increase in [Ca2+]i. RvD1-, but not the RvE1-, stimulated increase in [Ca2+]i and mucin secretion was blocked by the EGFR inhibitor AG1478 and siRNA for the EGFR. RvD1-, but not RvE1-stimulated an increase in [Ca2+]i that was also inhibited by TAPI-1, an inhibitor of the matrix metalloprotease ADAM 17. Inhibition of the EGFR also blocked RvD1-stimulated increase in AKT activity and both RvD1-and RvE1-stimulated increase in ERK 1/2 activity. Pretreatment with either RvD1 or RvE1 did not block the EGFR-stimulated increase in [Ca2+]i. CONCLUSIONS We conclude that in cultured rat and human conjunctival goblet cells, RvD1 activates the EGFR, increases [Ca2+]i, activates AKT and ERK1/2 to stimulate mucin secretion. RvE1 does not transactivate the EGFR to increase [Ca2+]I and stimulate mucin secretion, but does interact with the receptor to increase ERK 1/2 activity.
Collapse
Affiliation(s)
- Rebecca Kaye
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Nora Botten
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Marit Lippestad
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Dayu Li
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Robin R Hodges
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Tor P Utheim
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway; Department of Plastic and Reconstructive Surgery, University of Oslo, Oslo, Norway
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Darlene A Dartt
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
170
|
Tsuchida S, Matsuzaki T, Yamato M, Okuda K, Fu HY, Araki R, Sanada S, Asanuma H, Asano Y, Asakura M, Hao H, Takashima S, Kitakaze M, Sakata Y, Mekada E, Minamino T. Anti-HB-EGF Antibody-Mediated Delivery of siRNA to Atherosclerotic Lesions in Mice. Int Heart J 2018; 59:1425-1431. [DOI: 10.1536/ihj.17-644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Shota Tsuchida
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Takashi Matsuzaki
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University
| | - Masaki Yamato
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Keiji Okuda
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Hai Ying Fu
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Ryo Araki
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Shoji Sanada
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Hiroshi Asanuma
- Department of Internal Medicine, Meiji University of Integrative Medicine
| | - Yoshihiro Asano
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Masanori Asakura
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine
| | - Hiroyuki Hao
- Department of Pathology, Nihon University School of Medicine
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine
| | - Masafumi Kitakaze
- Clinical Research and Development, National Cerebral and Cardiovascular Center Research Institute
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Eisuke Mekada
- Department of Cell Biology, Research Institute for Microbial Diseases, Osaka University
| | - Tetsuo Minamino
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University
| |
Collapse
|
171
|
Trudel S, Lendvai N, Popat R, Voorhees PM, Reeves B, Libby EN, Richardson PG, Anderson LD, Sutherland HJ, Yong K, Hoos A, Gorczyca MM, Lahiri S, He Z, Austin DJ, Opalinska JB, Cohen AD. Targeting B-cell maturation antigen with GSK2857916 antibody-drug conjugate in relapsed or refractory multiple myeloma (BMA117159): a dose escalation and expansion phase 1 trial. Lancet Oncol 2018; 19:1641-1653. [PMID: 30442502 DOI: 10.1016/s1470-2045(18)30576-x] [Citation(s) in RCA: 196] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 07/17/2018] [Accepted: 07/24/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND B-cell maturation antigen (BCMA) is a cell-surface receptor of the tumour necrosis superfamily required for plasma cell survival. BMCA is universally detected on patient-derived myeloma cells and has emerged as a selective antigen to be targeted by novel treatments in multiple myeloma. We assessed the safety, tolerability, and preliminary clinical activity of GSK2857916, a novel anti-BCMA antibody conjugated to microtubule-disrupting agent monomethyl auristatin F, in patients with relapsed and refractory multiple myeloma. METHODS We did an international, multicentre, open-label, first-in-human phase 1 study with dose escalation (part 1) and dose expansion (part 2) phases, at nine centres in the USA, Canada, and the UK. Adults with histologically or cytologically confirmed multiple myeloma, Eastern Cooperative Oncology Group performance status 0 or 1, and progressive disease after stem cell transplantation, alkylators, proteasome inhibitors, and immunomodulators were recruited for this study. In part 1, patients received GSK2857916 (0·03-4·60 mg/kg) through 1 h intravenous infusions once every 3 weeks. In part 2, patients received the selected recommended phase 2 dose of GSK2857916 (3·40 mg/kg) once every 3 weeks. Primary endpoints were maximum tolerated dose and recommended phase 2 dose. Secondary endpoints for part 2 included preliminary anti-cancer clinical activity. All patients who received one or more doses were included in this prespecified administrative interim analysis (data cutoff date June 26, 2017), which was done for internal purposes. This study is registered with ClinicalTrials.gov, number NCT02064387, and is ongoing, but closed for recruitment. FINDINGS Between July 29, 2014, and Feb 21, 2017, we treated 73 patients: 38 patients in the dose-escalation part 1 and 35 patients in the dose-expansion part 2. There were no dose-limiting toxicities and no maximum tolerated dose was identified in part 1. On the basis of safety and clinical activity, we selected 3·40 mg/kg as the recommended phase 2 dose. Corneal events were common (20 [53%] of 38 patients in part 1 and 22 [63%] of 35 in part 2); most (18 [47%] in part 1 and 19 [54%] in part 2) were grade 1 or 2 and resulted in two treatment discontinuations in part 1 and no discontinuations in part 2. The most common grade 3 or 4 events were thrombocytopenia (13 [34%] of 38 patients in part 1 and 12 [34%] of 35 in part 2) and anaemia (6 [16%] in part 1 and 5 [14%] in part 2). There were 12 treatment-related serious adverse events and no treatment-related deaths. In part 2, 21 (60·0%; 95% CI 42·1-76·1) of 35 patients achieved an overall response. INTERPRETATION At the identified recommended phase 2 dose, GSK2857916 was well tolerated and had good clinical activity in heavily pretreated patients, thereby indicating that this might be a promising candidate for the treatment of relapsed or refractory multiple myeloma. FUNDING GlaxoSmithKline.
Collapse
Affiliation(s)
| | - Nikoletta Lendvai
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Medical College, Cornell University, New York, NY, USA
| | - Rakesh Popat
- NIHR University College London Hospital Clinical Research Facility, NHS Foundation Trust, London, UK
| | | | - Brandi Reeves
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | | | | | | | | | - Kwee Yong
- NIHR University College London Hospital Clinical Research Facility, NHS Foundation Trust, London, UK
| | - Axel Hoos
- GlaxoSmithKline, Philadelphia, PA, USA
| | | | | | | | | | | | - Adam D Cohen
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
172
|
Matulonis UA, Birrer MJ, O'Malley DM, Moore KN, Konner J, Gilbert L, Martin LP, Bauer TM, Oza AM, Malek K, Pinkas J, Kim SK. Evaluation of Prophylactic Corticosteroid Eye Drop Use in the Management of Corneal Abnormalities Induced by the Antibody–Drug Conjugate Mirvetuximab Soravtansine. Clin Cancer Res 2018; 25:1727-1736. [DOI: 10.1158/1078-0432.ccr-18-2474] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/01/2018] [Accepted: 11/06/2018] [Indexed: 11/16/2022]
|
173
|
|
174
|
Moore KN, O'Malley DM, Vergote I, Martin LP, Gonzalez-Martin A, Malek K, Birrer MJ. Safety and activity findings from a phase 1b escalation study of mirvetuximab soravtansine, a folate receptor alpha (FRα)-targeting antibody-drug conjugate (ADC), in combination with carboplatin in patients with platinum-sensitive ovarian cancer. Gynecol Oncol 2018; 151:46-52. [DOI: 10.1016/j.ygyno.2018.07.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/18/2018] [Accepted: 07/21/2018] [Indexed: 02/06/2023]
|
175
|
Richardson DL, Seward SM, Moore KN. Antibody Drug Conjugates in the Treatment of Epithelial Ovarian Cancer. Hematol Oncol Clin North Am 2018; 32:1057-1071. [PMID: 30390760 DOI: 10.1016/j.hoc.2018.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Antibody drug conjugates are novel mechanisms for delivering chemotherapy. They vary based on the targeted antigen, conjugated cytotoxic, and the type of linker used. These differences determine what cells are targeted. There are 2 antibody drug conjugates approved for use in cancer. For epithelial ovarian cancer, more than 15 antibody drug conjugates are under study. Using antibody drug conjugates in epithelial ovarian cancer makes sense. This review discusses promising trial results demonstrating efficacy. Reported toxicities include visual disturbance. There is an absence of significant hematologic toxicity. Overlapping toxicity between standard cytotoxics and antibody drug conjugates includes neuropathy and constitutional symptoms.
Collapse
Affiliation(s)
- Debra L Richardson
- Division of Gynecologic Oncology, Stephenson Oklahoma Cancer Institute, The University of Oklahoma, 800 Northeast 10th Street, Oklahoma City, OK 73104, USA.
| | - Shelly M Seward
- Division of Gynecologic Oncology, Karmanos Cancer Institute, Wayne State University, 4100 John R Street, Harper Professional Building, Suite 721, Detroit, MI 48070, USA
| | - Kathleen N Moore
- Division of Gynecologic Oncology, Stephenson Oklahoma Cancer Institute, The University of Oklahoma, 800 Northeast 10th Street, Oklahoma City, OK 73104, USA
| |
Collapse
|
176
|
Abstract
PURPOSE OF REVIEW Antibody-drug conjugates (ADCs) represent a promising new class of cancer therapeutics. Currently more than 60 ADCs are in clinical development, however, only very few trials focus on gynecologic malignancies. In this review, we summarize the most recent advances in ADC drug development with an emphasis on how this progress relates to patients diagnosed with gynecologic malignancies and breast cancer. RECENT FINDINGS The cytotoxic payloads of the majority of the ADCs that are currently in clinical trials for gynecologic malignancies or breast cancer are auristatins (MMAE, MMAF), maytansinoids (DM1, DM4), calicheamicin, pyrrolobenzodiazepines and SN-38. Both cleavable and noncleavable linkers are currently being investigated in clinical trials. A number of novel target antigens are currently being validated in ongoing clinical trials including folate receptor alpha, mesothelin, CA-125, NaPi2b, NOTCH3, protein tyrosine kinase-like 7, ephrin-A4, TROP2, CEACAM5, and LAMP1. For most ADCs currently in clinical development, dose-limiting toxicities appear to be unrelated to the targeted antigen but more tightly associated with the payload. Rational drug design involving optimization of the antibody, the linker and the conjugation chemistry is aimed at improving the therapeutic index of new ADCs. SUMMARY Antibody-drug conjugates can increase the efficacy and decrease the toxicity of their payloads in comparison with traditional cyctotoxic agents. A better and quicker translation of recent scientific advances in the field of ADCs into rational clinical trials for patients diagnosed with ovarian, endometrial or cervical cancer could create real improvements in tumor response, survival and quality of life for our patients.
Collapse
|
177
|
Watkins MP, Bartlett NL. CD19-targeted immunotherapies for treatment of patients with non-Hodgkin B-cell lymphomas. Expert Opin Investig Drugs 2018; 27:601-611. [PMID: 29940805 DOI: 10.1080/13543784.2018.1492549] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Ubiquitous expression of CD19 on B cell non-Hodgkin lymphoma identified it as a potential target for immune-based therapies. AREAS COVERED This article reviews the current literature on anti-CD19 therapies currently in clinical trials including monoclonal antibodies (mAb), antibody targeted cytotoxic drug conjugates (ADC), bispecific antibodies, and chimeric antigen receptor (CAR) modified T cells. EXPERT OPINION Naked anti-CD19 mAbs have shown little clinical benefit in B cell lymphomas. Despite unusual toxicity profiles with many anti-CD19 ADCs slowing development, durable remissions in a substantial minority of patients with refractory aggressive lymphomas should encourage continued efforts in this area. Blinatumomab, an anti-CD19 bispecific T cell engager, has shown impressive responses in relapse/refractory diffuse large B cell lymphoma (DLBCL), but is plagued by neurotoxicity issues and the need for continuous infusion. CD19 targeting CAR-T cell therapies are the most promising, with the potential for curing a third of refractory DLBCL patients. There is still much work to be done to address potentially life-threatening cytokine release syndrome and neurotoxicity, an extended production time precluding patients with rapidly progressive disease, and treatment expense. However, if the promise of CAR-T cell technology is confirmed, this will likely change the approach and prognosis for relapse/refractory aggressive lymphoma.
Collapse
|
178
|
Moore KN, Vergote I, Oaknin A, Colombo N, Banerjee S, Oza A, Pautier P, Malek K, Birrer MJ. FORWARD I: a Phase III study of mirvetuximab soravtansine versus chemotherapy in platinum-resistant ovarian cancer. Future Oncol 2018; 14:1669-1678. [DOI: 10.2217/fon-2017-0646] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Mirvetuximab soravtansine, an antibody–drug conjugate that binds with high affinity to folate receptor-α to provide tumor-directed delivery of the potent microtubule-disrupting agent DM4, has emerged as a promising investigational agent for the treatment of ovarian cancer, particularly in the setting of platinum-resistant disease. Here we describe the rationale and design of FORWARD I (NCT02631876), the first randomized, multicenter Phase III study to compare the safety and efficacy of mirvetuximab soravtansine versus investigator's choice of chemotherapy in women with folate receptor-α-positive, platinum-resistant epithelial ovarian, primary peritoneal or fallopian tube cancer. Patients will be randomized in a 2:1 ratio. The primary end point is progression-free survival, and key secondary objectives include comparison of overall response rates, overall survival and duration of response.
Collapse
Affiliation(s)
- Kathleen N Moore
- Department of Obstetrics & Gynecology, Stephenson Oklahoma Cancer Center at the University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Ignace Vergote
- Gynaecological Oncology, Leuven Cancer Institute, Leuven 3000, Belgium
| | - Ana Oaknin
- Medical Oncology Department, Vall D'Hebron Institute of Oncology (VHIO), Barcelona 08035, Spain
| | - Nicoletta Colombo
- Gynecologic Oncology, The European Institute of Oncology, Milan 20141, Italy
| | - Susana Banerjee
- Gynaecology Unit, Royal Marsden Hospital, London, SW3 6JJ, UK
| | - Amit Oza
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto M5G 2M9, Canada
| | - Patricia Pautier
- Department of Adult Medicine, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Karim Malek
- Clinical Development, ImmunoGen, Inc., Waltham, MA 02451, USA
| | - Michael J Birrer
- Division of Hematology–Oncology, University of Alabama at Birmingham Comprehensive Cancer Center, Birmingham, AL 35294, USA
| |
Collapse
|
179
|
Thompson JA, Motzer RJ, Molina AM, Choueiri TK, Heath EI, Redman BG, Sangha RS, Ernst DS, Pili R, Kim SK, Reyno L, Wiseman A, Trave F, Anand B, Morrison K, Doñate F, Kollmannsberger CK. Phase I Trials of Anti-ENPP3 Antibody-Drug Conjugates in Advanced Refractory Renal Cell Carcinomas. Clin Cancer Res 2018; 24:4399-4406. [PMID: 29848572 DOI: 10.1158/1078-0432.ccr-18-0481] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/19/2018] [Accepted: 05/22/2018] [Indexed: 01/02/2023]
Abstract
Purpose: To determine the safety, pharmacokinetics, and recommended phase II dose of an antibody-drug conjugate (ADC) targeting ectonucleotide phosphodiesterases-pyrophosphatase 3 (ENPP3) conjugated to monomethyl auristatin F (MMAF) in subjects with advanced metastatic renal cell carcinoma (mRCC).Patients and Methods: Two phase I studies were conducted sequentially with 2 ADCs considered equivalent, hybridoma-derived AGS-16M8F and Chinese hamster ovary-derived AGS-16C3F. AGS-16M8F was administered intravenously every 3 weeks at 5 dose levels ranging from 0.6 to 4.8 mg/kg until unacceptable toxicity or progression. The study was terminated before reaching the MTD. A second study with AGS-16C3F started with the AGS-16M8F bridging dose of 4.8 mg/kg given every 3 weeks.Results: The AGS-16M8F study (n = 26) closed before reaching the MTD. The median duration of treatment was 12 weeks (1.7-83 weeks). One subject had durable partial response (PR; 83 weeks) and 1 subject had prolonged stable disease (48 weeks). In the AGS-16C3F study (n = 34), the protocol-defined MTD was 3.6 mg/kg, but this was not tolerated in multiple doses. Reversible keratopathy was dose limiting and required multiple dose deescalations. The 1.8 mg/kg dose was determined to be safe and was associated with clinically relevant signs of antitumor response. Three of 13 subjects at 1.8 mg/kg had durable PRs (range, 100-143 weeks). Eight subjects at 2.7 mg/kg and 1.8 mg/kg had disease control >37 weeks (37.5-141 weeks).Conclusions: AGS-16C3F was tolerated and had durable antitumor activity at 1.8 mg/kg every 3 weeks. Clin Cancer Res; 24(18); 4399-406. ©2018 AACR.
Collapse
Affiliation(s)
- John A Thompson
- Division of Medical Oncology, University of Washington, Seattle, Washington.
| | - Robert J Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ana M Molina
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York, New York
| | - Toni K Choueiri
- Harvard Medical School, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Elisabeth I Heath
- Division of Hematology/Oncology, Karmanos Cancer Center, Detroit, Michigan
| | - Bruce G Redman
- Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan
| | | | - D Scott Ernst
- London Health Sciences Centre, London, Ontario, Canada
| | - Roberto Pili
- Department of Oncology, Indiana University, Bloomington, Indiana
| | - Stella K Kim
- Department of Opthalmology and Visual Science, University of Texas McGovern Medical School, Houston, Texas
| | - Leonard Reyno
- Department of Translational Research, Agensys, Inc. Santa Monica, California
| | - Aya Wiseman
- Department of Translational Research, Agensys, Inc. Santa Monica, California
| | | | | | - Karen Morrison
- Department of Translational Research, Agensys, Inc. Santa Monica, California
| | - Fernando Doñate
- Department of Translational Research, Agensys, Inc. Santa Monica, California
| | | |
Collapse
|
180
|
Goss GD, Vokes EE, Gordon MS, Gandhi L, Papadopoulos KP, Rasco DW, Fischer JS, Chu KL, Ames WW, Mittapalli RK, Lee HJ, Zeng J, Roberts-Rapp LA, Loberg LI, Ansell PJ, Reilly EB, Ocampo CJ, Holen KD, Tolcher AW. Efficacy and safety results of depatuxizumab mafodotin (ABT-414) in patients with advanced solid tumors likely to overexpress epidermal growth factor receptor. Cancer 2018. [PMID: 29533458 PMCID: PMC5969257 DOI: 10.1002/cncr.31304] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) alterations are associated with multiple cancers. Current EGFR-directed therapies have led to increased efficacy but are associated with specific side effects. The antibody-drug conjugate depatuxizumab mafodotin (depatux-m) targets EGFR with a monoclonal antibody linked to a cytotoxin, and is highly tumor-specific. METHODS This phase 1/2 study evaluated the safety, pharmacokinetics, and efficacy of depatux-m in patients who had advanced solid tumors with known wild-type EGFR overexpression, amplification, or mutated EGFR variant III. A 3 + 3 dose escalation was used, and 2 dosing schedules were evaluated. Depatux-m also was manufactured under an alternate process to reduce the drug load and improve the safety profile, and it was tested at the maximum tolerated dose (MTD). In another cohort, prolonged infusion time of depatux-m was evaluated; and a cohort with confirmed EGFR amplification also was evaluated at the MTD. RESULTS Fifty-six patients were treated. The MTD and the recommended phase 2 dose for depatux-m was 3.0 mg/kg. Common adverse events (AEs) were blurred vision (48%) and fatigue (41%). A majority of patients (66%) experienced 1 or more ocular AEs. Grade 3 or 4 AEs were observed in 43% of patients. One patient with EGFR-amplified, triple-negative breast cancer had a partial response. Stable disease was observed in 23% of patients. Pharmacokinetics revealed that depatux-m exposures were approximately dose-proportional. CONCLUSIONS Depatux-m resulted in infrequent nonocular AEs but increased ocular AEs. Patient follow-up confirmed that ocular AEs were reversible. Lowering the drug-antibody ratio did not decrease the number of ocular AEs. A partial response in 1 patient with EGFR-amplified disease provides the opportunity to study depatux-m in diseases with a high incidence of EGFR amplification. Cancer 2018;124:2174-83. © 2018 The Authors. Cancer published by Wiley Periodicals, Inc. on behalf of American Cancer Society. This is an open access article under the terms of the Creative Commons Attribution NonCommercial License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes.
Collapse
Affiliation(s)
- Glenwood D Goss
- The Ottawa Hospital Research Institute and the University of Ottawa, Ottawa, Ontario, Canada
| | - Everett E Vokes
- Department of Medicine, University of Chicago, Chicago, Illinois
| | | | - Leena Gandhi
- New York University Perlmutter Cancer Center, NYU Health, New York, New York
| | | | - Drew W Rasco
- South Texas Accelerated Research Therapeutics, San Antonio, Texas
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Kreps EO, Derveaux T, Denys H. Corneal Changes in Trastuzumab Emtansine Treatment. Clin Breast Cancer 2018; 18:e427-e429. [PMID: 29615304 DOI: 10.1016/j.clbc.2018.03.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 02/22/2018] [Accepted: 03/06/2018] [Indexed: 11/25/2022]
Affiliation(s)
- Elke O Kreps
- Department of Ophthalmology, Ghent University Hospitals, Ghent, Belgium.
| | - Thierry Derveaux
- Department of Ophthalmology, Ghent University Hospitals, Ghent, Belgium
| | - Hannelore Denys
- Department of Medical Oncology, Ghent University Hospitals, Ghent, Belgium
| |
Collapse
|
182
|
Phillips AC, Boghaert ER, Vaidya KS, Falls HD, Mitten MJ, DeVries PJ, Benatuil L, Hsieh CM, Meulbroek JA, Panchal SC, Buchanan FG, Durbin KR, Voorbach MJ, Reuter DR, Mudd SR, Loberg LI, Ralston SL, Cao D, Gan HK, Scott AM, Reilly EB. Characterization of ABBV-221, a Tumor-Selective EGFR-Targeting Antibody Drug Conjugate. Mol Cancer Ther 2018; 17:795-805. [PMID: 29483208 DOI: 10.1158/1535-7163.mct-17-0710] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 12/07/2017] [Accepted: 02/06/2018] [Indexed: 11/16/2022]
Abstract
Depatuxizumab mafodotin (depatux-m, ABT-414) is a tumor-selective antibody drug conjugate (ADC) comprised of the anti-EGFR antibody ABT-806 and the monomethyl auristatin F (MMAF) warhead. Depatux-m has demonstrated promising clinical activity in glioblastoma multiforme (GBM) patients and is currently being evaluated in clinical trials in first-line and recurrent GBM disease settings. Depatux-m responses have been restricted to patients with amplified EGFR, highlighting the need for therapies with activity against tumors with nonamplified EGFR overexpression. In addition, depatux-m dosing has been limited by corneal side effects common to MMAF conjugates. We hypothesized that a monomethyl auristatin E (MMAE) ADC utilizing an EGFR-targeting antibody with increased affinity may have broader utility against tumors with more modest EGFR overexpression while mitigating the risk of corneal side effects. We describe here preclinical characterization of ABBV-221, an EGFR-targeting ADC comprised of an affinity-matured ABT-806 conjugated to MMAE. ABBV-221 binds to a similar EGFR epitope as depatux-m and retains tumor selectivity with increased binding to EGFR-positive tumor cells and greater in vitro potency. ABBV-221 displays increased tumor uptake and antitumor activity against wild-type EGFR-positive xenografts with a greatly reduced incidence of corneal side effects relative to depatux-m. ABBV-221 has similar activity as depatux-m against an EGFR-amplified GBM patient derived xenograft (PDX) model and is highly effective alone and in combination with standard-of-care temozolomide in an EGFRvIII-positive GBM xenograft model. Based on these results, ABBV-221 has advanced to a phase I clinical trial in patients with advanced solid tumors associated with elevated levels of EGFR. Mol Cancer Ther; 17(4); 795-805. ©2018 AACR.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Diana Cao
- Olivia Newton-John Cancer Research Institute, and La Trobe University Austin Hospital, Heidelberg, Victoria, Australia
| | - Hui K Gan
- Olivia Newton-John Cancer Research Institute, and La Trobe University Austin Hospital, Heidelberg, Victoria, Australia
| | - Andrew M Scott
- Olivia Newton-John Cancer Research Institute, and La Trobe University Austin Hospital, Heidelberg, Victoria, Australia
| | | |
Collapse
|
183
|
Modulation of Macropinocytosis-Mediated Internalization Decreases Ocular Toxicity of Antibody–Drug Conjugates. Cancer Res 2018; 78:2115-2126. [DOI: 10.1158/0008-5472.can-17-3202] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 12/19/2017] [Accepted: 01/25/2018] [Indexed: 11/16/2022]
|
184
|
Moore KN, Martin LP, O'Malley DM, Matulonis UA, Konner JA, Vergote I, Ponte JF, Birrer MJ. A review of mirvetuximab soravtansine in the treatment of platinum-resistant ovarian cancer. Future Oncol 2018; 14:123-136. [DOI: 10.2217/fon-2017-0379] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Resistance to platinum-based therapy poses a significant clinical challenge for the management of advanced ovarian cancer, a leading cause of cancer mortality among women. Mirvetuximab soravtansine is a novel antibody–drug conjugate that targets folate receptor-α, a validated molecular target for therapeutic intervention in this disease. Here, we examine mirvetuximab soravtansine's mechanism of action and pharmacology, and review its clinical evaluation in ovarian cancer to date. We focus on the favorable tolerability and encouraging signals of efficacy that have emerged, most notably in patients with platinum-resistant disease. Ongoing Phase III monotherapy and Phase Ib/II combination trials evaluating its activity in the setting of platinum resistance are emphasized, which will help define its role in the evolving landscape of ovarian cancer therapy.
Collapse
Affiliation(s)
- Kathleen N Moore
- Department of Obstetrics & Gynecology, Stephenson Oklahoma Cancer Center at the University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Lainie P Martin
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - David M O'Malley
- Department of Obstetrics & Gynecology, The Ohio State University, Columbus, OH 43210, USA
| | - Ursula A Matulonis
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02115, USA
| | - Jason A Konner
- Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ignace Vergote
- Gynaecological Oncology, Leuven Cancer Institute, Leuven 3000, Belgium
| | - Jose F Ponte
- Pharmacology, ImmunoGen, Inc, Waltham, MA 02451, USA
| | - Michael J Birrer
- Gillette Center for Gynecologic Oncology, Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
185
|
Guffroy M, Falahatpisheh H, Finkelstein M. Improving the Safety Profile of ADCs. CANCER DRUG DISCOVERY AND DEVELOPMENT 2018. [DOI: 10.1007/978-3-319-78154-9_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
186
|
van den Bent M, Gan HK, Lassman AB, Kumthekar P, Merrell R, Butowski N, Lwin Z, Mikkelsen T, Nabors LB, Papadopoulos KP, Penas-Prado M, Simes J, Wheeler H, Walbert T, Scott AM, Gomez E, Lee HJ, Roberts-Rapp L, Xiong H, Bain E, Ansell PJ, Holen KD, Maag D, Reardon DA. Efficacy of depatuxizumab mafodotin (ABT-414) monotherapy in patients with EGFR-amplified, recurrent glioblastoma: results from a multi-center, international study. Cancer Chemother Pharmacol 2017; 80:1209-1217. [PMID: 29075855 PMCID: PMC5686264 DOI: 10.1007/s00280-017-3451-1] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 10/07/2017] [Indexed: 11/06/2022]
Abstract
PURPOSE Patients with recurrent glioblastoma (rGBM) have a poor prognosis. Epidermal growth factor receptor (EGFR) gene amplification is present in ~ 50% of glioblastomas (GBMs). Depatuxizumab mafodotin (depatux-m), formerly ABT-414, is an antibody-drug conjugate that preferentially binds cells with EGFR amplification, is internalized and releases a potent antimicrotubule agent, monomethyl auristatin F (MMAF). Here we report the safety, pharmacokinetics, and efficacy of depatux-m monotherapy at the recommended Phase 2 dose (RPTD) in patients with EGFR-amplified, rGBM. METHODS M12-356 (NCT01800695) is an open-label study with three escalation and expansion cohorts. Sixty-six patients with EGFR-amplified, rGBM were treated with depatux-m monotherapy at 1.25 mg/kg intravenously every 2 weeks. Adults with measurable rGBM, who were bevacizumab-naïve, with EGFR amplification were eligible. RESULTS Among 66 patients, median age was 58 years (range 35-80). All patients were previously treated with radiotherapy/temozolomide. The most common adverse events (AEs) were eye related (91%), including blurred vision (65%), dry eye (29%), keratitis, and photophobia (27% each). Grade 3/4 AEs occurred in 42% of all patients, and ocular Grade 3/4 AEs occurred in 33% of patients overall. One patient (2%) had a Grade 4 ocular AE. Ocular AEs were manageable and usually resolved once treatment with depatux-m ceased. The objective response rate was 6.8%, the 6-month progression-free survival rate was 28.8%, and the 6-month overall survival rate was 72.5%. CONCLUSION Depatux-m monotherapy displayed frequent but mostly Grade 1/2 ocular toxicities. A PFS6 of 28.8% was observed in this rGBM population, warranting further study.
Collapse
Affiliation(s)
- Martin van den Bent
- Brain Tumor Center, Erasmus MC Cancer Institute, Groene Hilledijk 301, 3075 EA, Rotterdam, the Netherlands.
| | - Hui K Gan
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Andrew B Lassman
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
| | | | - Ryan Merrell
- NorthShore University Health System, Evanston, IL, USA
| | - Nicholas Butowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Zarnie Lwin
- Department of Medical Oncology, School of Medicine, University of Queensland, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | | | - Louis B Nabors
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | - John Simes
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Helen Wheeler
- Medical Oncology, Royal North Shore Hospital, Sydney, NSW, Australia
| | | | - Andrew M Scott
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
187
|
Ryan MC, Palanca-Wessels MC, Schimpf B, Gordon KA, Kostner H, Meyer B, Yu C, Van Epps HA, Benjamin D. Therapeutic potential of SGN-CD19B, a PBD-based anti-CD19 drug conjugate, for treatment of B-cell malignancies. Blood 2017; 130:2018-2026. [PMID: 28903943 PMCID: PMC5669207 DOI: 10.1182/blood-2017-04-779389] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 08/28/2017] [Indexed: 12/11/2022] Open
Abstract
Patients with relapsed/refractory B-cell malignancies such as non-Hodgkin lymphoma (B-NHL) or acute lymphoblastic leukemia have a poor prognosis. Despite measurable clinical activity with new targeted therapies, many patients do not achieve a complete or durable response suggesting an opportunity to improve upon existing therapies. Here we describe SGN-CD19B, a pyrrolobenzodiazepine (PBD)-based anti-CD19 antibody drug conjugate (ADC) being investigated for treatment of B-cell malignancies, which has improved potency compared with other ADCs. CD19-expressing tumor cells rapidly internalize SGN-CD19B, and the released PBD drug induces DNA damage, resulting in G2/M cell cycle arrest and cell death. SGN-CD19B demonstrated activity against a broad panel of malignant B-cell lines and induced durable regressions in mice bearing xenografts derived from these B-cell malignancies. A single dose of SGN-CD19B induced durable regressions at 300 μg/kg (3 μg/kg drug equivalents); combination with rituximab decreased the curative dose to 100 μg/kg (1 μg/kg drug equivalents). These doses are significantly lower than the level of drug required with other ADC payloads. In cynomolgus monkeys, SGN-CD19B effectively depleted CD20+ B lymphocytes in peripheral blood and lymphoid tissues confirming that SGN-CD19B is pharmacodynamically active at well-tolerated doses. In summary, preclinical studies show SGN-CD19B is a highly active ADC, which releases a DNA cross-linking agent rather than a microtubule inhibitor. The distinct mechanism of action, broad potency, and potential to combine with rituximab suggest that SGN-CD19B may offer unique clinical opportunities in B-cell malignancies. A phase 1 clinical trial is in progress to investigate the therapeutic potential of SGN-CD19B in relapsed/refractory B-NHL. This trial was registered at www.clinicaltrials.gov as #NCT02702141.
Collapse
Affiliation(s)
| | | | | | | | | | - Brad Meyer
- Department of Translational Research and
| | - Changpu Yu
- Department of Translational Research and
| | | | | |
Collapse
|
188
|
Martin LP, Konner JA, Moore KN, Seward SM, Matulonis UA, Perez RP, Su Y, Berkenblit A, Ruiz-Soto R, Birrer MJ. Characterization of folate receptor alpha (FRα) expression in archival tumor and biopsy samples from relapsed epithelial ovarian cancer patients: A phase I expansion study of the FRα-targeting antibody-drug conjugate mirvetuximab soravtansine. Gynecol Oncol 2017; 147:402-407. [PMID: 28843653 PMCID: PMC6893864 DOI: 10.1016/j.ygyno.2017.08.015] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/08/2017] [Accepted: 08/12/2017] [Indexed: 11/30/2022]
Abstract
PURPOSE To characterize folate receptor alpha (FRα) expression in archival and fresh biopsy tumor samples from relapsed ovarian cancer patients. METHODS Patients with ovarian tumors amenable to biopsy were eligible to enroll. Eligibility included a minimum requirement of FRα positivity in archival tumor samples (≥25% of cells with ≥2+ staining intensity). Patients received mirvetuximab soravtansine at 6mg/kg once every 3weeks. Core needle biopsies were collected before and after treatment and FRα levels assessed by immunohistochemistry. Descriptive statistics were used to summarize the association between receptor expression and response. RESULTS Twenty-seven heavily pre-treated patients were enrolled. Six individuals (22%) did not have evaluable pre-treatment biopsies due to insufficient tumor cells. The concordance of FRα expression in archival and biopsy tissues was 71%, and no major shifts in receptor expression were seen in matched pre- and post-treatment biopsy samples. Adverse events were generally mild (≤grade 2) with keratopathy (48%), fatigue (44%), diarrhea, and blurred vision (each 37%) being the most common treatment-related toxicities. The confirmed objective response rate (ORR) was 22%, including two complete responses and four partial responses. Superior efficacy measures were observed in the subset of patients with the highest FRα levels (ORR, 31%; progression-free survival, 5.4months). CONCLUSION Concordance of FRα expression in biopsy versus archival tumor samples suggests that archival tissue can reliably identify patients with receptor-positive tumors and is appropriate for patient selection in mirvetuximab soravtansine clinical trials. Regardless of the tissue source analyzed, higher FRα expression was associated with greater antitumor activity.
Collapse
Affiliation(s)
| | - Jason A Konner
- Memorial Sloan Kettering Cancer Center, New York, NY, United States.
| | - Kathleen N Moore
- Stephenson Oklahoma Cancer Center at the University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.
| | - Shelly M Seward
- Wayne State University - Karmanos Cancer Institute, Huntington Woods, MI, United States.
| | | | | | - Ying Su
- ImmunoGen, Inc., Waltham, MA, United States.
| | | | | | | |
Collapse
|
189
|
Shapiro GI, Vaishampayan UN, LoRusso P, Barton J, Hua S, Reich SD, Shazer R, Taylor CT, Xuan D, Borghaei H. First-in-human trial of an anti-5T4 antibody-monomethylauristatin conjugate, PF-06263507, in patients with advanced solid tumors. Invest New Drugs 2017; 35:315-323. [PMID: 28070718 PMCID: PMC5418317 DOI: 10.1007/s10637-016-0419-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 12/22/2016] [Indexed: 12/20/2022]
Abstract
Background The antibody-drug conjugate PF-06263507 targets the cell-surface, tumor-associated antigen 5T4 and consists of a humanized IgG1 conjugated to the microtubule-disrupting agent monomethylauristatin-F by a non-cleavable maleimidocaproyl linker. In this first-in-human, dose-finding trial (NCT01891669), we evaluated safety, pharmacokinetics, and preliminary antitumor activity of PF-06263507 in pretreated patients with advanced solid tumors, unselected for 5T4 expression. starting at 0.05 mg/kg, with 25, 56, and 95% dose increments, depending on observed dose-limiting toxicities (DLTs), applying a modified continual reassessment method. Results Twenty-six patients received PF-06263507 at 0.05 to 6.5 mg/kg. The first DLT, grade 3 photophobia, occurred at 4.34 mg/kg and two additional DLTs, grade 2 keratitis and grade 1 limbal stem cell deficiency (> 2-week dosing delay), at 6.5 mg/kg. The most common adverse events (AEs) were fatigue (38.5%), photophobia (26.9%), and decreased appetite, dry eye, nausea, and thrombocytopenia (23.1% each). No treatment-related grade 4-5 AEs were reported. Systemic exposure of PF-06263507 increased in a dose-related manner. At the maximum tolerated dose (MTD, 4.34 mg/kg), mean terminal half-life for PF-06263507 and unconjugated payload were ~6 and 3 days, respectively. Payload serum concentrations were substantially lower compared with PF-06263507. No objective responses were observed. Conclusions The MTD and recommended phase II dose were determined to be 4.34 mg/kg. Ocular toxicities accounted for the DLTs observed, as previously reported with monomethylauristatin-F payloads. Further studies are warranted to investigate clinical activity of this agent in patients with 5T4-expressing tumors.Trial registration ID: NCT01891669.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antigens, Neoplasm/adverse effects
- Antigens, Neoplasm/therapeutic use
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/pharmacokinetics
- Antineoplastic Agents, Immunological/therapeutic use
- Female
- Humans
- Immunoconjugates/adverse effects
- Immunoconjugates/pharmacokinetics
- Immunoconjugates/therapeutic use
- Keratitis/chemically induced
- Male
- Maximum Tolerated Dose
- Membrane Glycoproteins/antagonists & inhibitors
- Middle Aged
- Neoplasms/drug therapy
- Neoplasms/metabolism
- Oligopeptides/adverse effects
- Oligopeptides/pharmacokinetics
- Oligopeptides/therapeutic use
- Photophobia/chemically induced
- Treatment Outcome
Collapse
Affiliation(s)
- Geoffrey I Shapiro
- Early Drug Development Center, Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Mayer 446, Boston, MA, 02215, USA.
| | | | | | | | | | | | | | - Carrie T Taylor
- Pfizer Oncology, La Jolla, CA, USA.
- Pfizer Early Oncology Development and Clinical Research, 10777 Science Center Drive, CB-1, San Diego, CA, 92121, USA.
| | | | | |
Collapse
|
190
|
Bialucha CU, Collins SD, Li X, Saxena P, Zhang X, Dürr C, Lafont B, Prieur P, Shim Y, Mosher R, Lee D, Ostrom L, Hu T, Bilic S, Rajlic IL, Capka V, Jiang W, Wagner JP, Elliott G, Veloso A, Piel JC, Flaherty MM, Mansfield KG, Meseck EK, Rubic-Schneider T, London AS, Tschantz WR, Kurz M, Nguyen D, Bourret A, Meyer MJ, Faris JE, Janatpour MJ, Chan VW, Yoder NC, Catcott KC, McShea MA, Sun X, Gao H, Williams J, Hofmann F, Engelman JA, Ettenberg SA, Sellers WR, Lees E. Discovery and Optimization of HKT288, a Cadherin-6-Targeting ADC for the Treatment of Ovarian and Renal Cancers. Cancer Discov 2017; 7:1030-1045. [PMID: 28526733 DOI: 10.1158/2159-8290.cd-16-1414] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/11/2017] [Accepted: 05/10/2017] [Indexed: 11/16/2022]
Abstract
Despite an improving therapeutic landscape, significant challenges remain in treating the majority of patients with advanced ovarian or renal cancer. We identified the cell-cell adhesion molecule cadherin-6 (CDH6) as a lineage gene having significant differential expression in ovarian and kidney cancers. HKT288 is an optimized CDH6-targeting DM4-based antibody-drug conjugate (ADC) developed for the treatment of these diseases. Our study provides mechanistic evidence supporting the importance of linker choice for optimal antitumor activity and highlights CDH6 as an antigen for biotherapeutic development. To more robustly predict patient benefit of targeting CDH6, we incorporate a population-based patient-derived xenograft (PDX) clinical trial (PCT) to capture the heterogeneity of response across an unselected cohort of 30 models-a novel preclinical approach in ADC development. HKT288 induces durable tumor regressions of ovarian and renal cancer models in vivo, including 40% of models on the PCT, and features a preclinical safety profile supportive of progression toward clinical evaluation.Significance: We identify CDH6 as a target for biotherapeutics development and demonstrate how an integrated pharmacology strategy that incorporates mechanistic pharmacodynamics and toxicology studies provides a rich dataset for optimizing the therapeutic format. We highlight how a population-based PDX clinical trial and retrospective biomarker analysis can provide correlates of activity and response to guide initial patient selection for first-in-human trials of HKT288. Cancer Discov; 7(9); 1030-45. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 920.
Collapse
Affiliation(s)
- Carl U Bialucha
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts.
| | - Scott D Collins
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Xiao Li
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Parmita Saxena
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Xiamei Zhang
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Clemens Dürr
- Novartis Institutes for Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Bruno Lafont
- Novartis Institutes for Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Pierric Prieur
- Novartis Institutes for Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Yeonju Shim
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Rebecca Mosher
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - David Lee
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Lance Ostrom
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Tiancen Hu
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Sanela Bilic
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | | | - Vladimir Capka
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Wei Jiang
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Joel P Wagner
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - GiNell Elliott
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Artur Veloso
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Jessica C Piel
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Meghan M Flaherty
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Keith G Mansfield
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Emily K Meseck
- Novartis Institutes for Biomedical Research, East Hanover, New Jersey
| | - Tina Rubic-Schneider
- Novartis Institutes for Biomedical Research, Campus Klybeckstrasse, Basel, Switzerland
| | | | | | - Markus Kurz
- Novartis Pharma AG, Novartis Campus, Basel, Switzerland
| | - Duc Nguyen
- Novartis Pharma, Cambridge, Massachusetts
| | - Aaron Bourret
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Matthew J Meyer
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Jason E Faris
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Mary J Janatpour
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Vivien W Chan
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | | | | | | | | | - Hui Gao
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Juliet Williams
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Francesco Hofmann
- Novartis Institutes for Biomedical Research, Campus Klybeckstrasse, Basel, Switzerland
| | | | - Seth A Ettenberg
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - William R Sellers
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Emma Lees
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| |
Collapse
|
191
|
Drug-induced corneal epithelial changes. Surv Ophthalmol 2017; 62:286-301. [DOI: 10.1016/j.survophthal.2016.11.008] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 11/15/2016] [Accepted: 11/18/2016] [Indexed: 11/20/2022]
|
192
|
Moore KN, Borghaei H, O'Malley DM, Jeong W, Seward SM, Bauer TM, Perez RP, Matulonis UA, Running KL, Zhang X, Ponte JF, Ruiz-Soto R, Birrer MJ. Phase 1 dose-escalation study of mirvetuximab soravtansine (IMGN853), a folate receptor α-targeting antibody-drug conjugate, in patients with solid tumors. Cancer 2017; 123:3080-3087. [PMID: 28440955 DOI: 10.1002/cncr.30736] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 02/02/2017] [Accepted: 03/20/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Mirvetuximab soravtansine (IMGN853) is an antibody-drug conjugate that selectively targets folate receptor α (FRα). In this phase 1 dose-escalation study, the authors investigated IMGN853 in patients with FRα-positive solid tumors. METHODS Patients received IMGN853 on day 1 of a 21-day cycle (once every 3 weeks dosing), with cycles repeated until patients experienced dose-limiting toxicity or progression. Dose escalation commenced in single-patient cohorts for the first 4 planned dose levels and then followed a standard 3 + 3 scheme. The primary objectives were to determine the maximum tolerated dose and the recommended phase 2 dose. Secondary objectives were to determine safety and tolerability, to characterize the pharmacokinetic profile, and to describe preliminary clinical activity. RESULTS In total, 44 patients received treatment at doses escalating from 0.15 to 7.0 mg/kg. No meaningful drug accumulation was observed with the dosing regimen of once every 3 weeks. The most common treatment-related adverse events were fatigue, blurred vision, and diarrhea, the majority of which were grade 1 or 2. The dose-limiting toxicities observed were grade 3 hypophosphatemia (5.0 mg/kg) and grade 3 punctate keratitis (7.0 mg/kg). Two patients, both of whom were individuals with epithelial ovarian cancer, achieved confirmed tumor responses according to Response Evaluation Criteria in Solid Tumors 1.1, and each was a partial response. CONCLUSIONS IMGN853 demonstrated a manageable safety profile and encouraging preliminary clinical activity, particularly in patients with ovarian cancer. The results establish a recommended phase 2 dosing of 6.0 mg/kg (based on adjusted ideal body weight) once every 3 weeks. Cancer 2017. © 2017 American Cancer Society. Cancer 2017;123:3080-7. © 2017 American Cancer Society.
Collapse
Affiliation(s)
- Kathleen N Moore
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Hossein Borghaei
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - David M O'Malley
- Department of Obstetrics and Gynecology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Woondong Jeong
- Cancer Therapy and Research Center, University of Texas Health Science Center, San Antonio, Texas
| | - Shelly M Seward
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan
| | - Todd M Bauer
- Sarah Cannon Research Institute/Tennessee Oncology, PLLC, Nashville, Tennessee
| | - Raymond P Perez
- Division of Hematology/Oncology, University of Kansas, Fairway, Kansas
| | - Ursula A Matulonis
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | | | | | | | | | - Michael J Birrer
- Gillette Center for Gynecologic Oncology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
193
|
Moore KN, Martin LP, O’Malley DM, Matulonis UA, Konner JA, Perez RP, Bauer TM, Ruiz-Soto R, Birrer MJ. Safety and Activity of Mirvetuximab Soravtansine (IMGN853), a Folate Receptor Alpha-Targeting Antibody-Drug Conjugate, in Platinum-Resistant Ovarian, Fallopian Tube, or Primary Peritoneal Cancer: A Phase I Expansion Study. J Clin Oncol 2017; 35:1112-1118. [PMID: 28029313 PMCID: PMC5559878 DOI: 10.1200/jco.2016.69.9538] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose This phase I expansion cohort study evaluated the safety and clinical activity of mirvetuximab soravtansine (IMGN853), an antibody-drug conjugate consisting of a humanized anti-folate receptor alpha (FRα) monoclonal antibody linked to the tubulin-disrupting maytansinoid DM4, in a population of patients with FRα-positive and platinum-resistant ovarian cancer. Patients and Methods Patients with platinum-resistant epithelial ovarian, fallopian tube, or primary peritoneal cancer received IMGN853 at 6.0 mg/kg (adjusted ideal body weight) once every 3 weeks. Eligibility included a minimum requirement of FRα positivity by immunohistochemistry (≥ 25% of tumor cells with at least 2+ staining intensity). Adverse events, tumor response (via Response Evaluation Criteria in Solid Tumors [RECIST] version 1.1), and progression-free survival (PFS) were determined. Results Forty-six patients were enrolled. Adverse events were generally mild (≤ grade 2), with diarrhea (44%), blurred vision (41%), nausea (37%), and fatigue (30%) being the most commonly observed treatment-related toxicities. Grade 3 fatigue and hypotension were reported in two patients each (4%). For all evaluable patients, the confirmed objective response rate was 26%, including one complete and 11 partial responses, and the median PFS was 4.8 months. The median duration of response was 19.1 weeks. Notably, in the subset of patients who had received three or fewer prior lines of therapy (n = 23), an objective response rate of 39%, PFS of 6.7 months, and duration of response of 19.6 weeks were observed. Conclusion IMGN853 exhibited a manageable safety profile and was active in platinum-resistant ovarian cancer, with the strongest signals of efficacy observed in less heavily pretreated individuals. On the basis of these findings, the dose, schedule, and target population were identified for a phase III trial of IMGN853 monotherapy in patients with platinum-resistant disease.
Collapse
Affiliation(s)
- Kathleen N. Moore
- Kathleen N. Moore, University of Oklahoma Health Sciences Center, Oklahoma City, OK; Lainie P. Martin, Fox Chase Cancer Center, Philadelphia, PA; David M. O’Malley, Ohio State University James Cancer Center, Columbus, OH; Ursula A. Matulonis, Dana-Farber Cancer Institute; Michael J. Birrer, Massachusetts General Hospital, Boston; Rodrigo Ruiz-Soto, ImmunoGen, Waltham, MA; Jason A. Konner, Memorial Sloan Kettering Cancer Center, New York, NY; Raymond P. Perez, University of Kansas, Fairway, KS; and Todd M. Bauer, Sarah Cannon Research Institute and Tennessee Oncology, Nashville, TN
| | - Lainie P. Martin
- Kathleen N. Moore, University of Oklahoma Health Sciences Center, Oklahoma City, OK; Lainie P. Martin, Fox Chase Cancer Center, Philadelphia, PA; David M. O’Malley, Ohio State University James Cancer Center, Columbus, OH; Ursula A. Matulonis, Dana-Farber Cancer Institute; Michael J. Birrer, Massachusetts General Hospital, Boston; Rodrigo Ruiz-Soto, ImmunoGen, Waltham, MA; Jason A. Konner, Memorial Sloan Kettering Cancer Center, New York, NY; Raymond P. Perez, University of Kansas, Fairway, KS; and Todd M. Bauer, Sarah Cannon Research Institute and Tennessee Oncology, Nashville, TN
| | - David M. O’Malley
- Kathleen N. Moore, University of Oklahoma Health Sciences Center, Oklahoma City, OK; Lainie P. Martin, Fox Chase Cancer Center, Philadelphia, PA; David M. O’Malley, Ohio State University James Cancer Center, Columbus, OH; Ursula A. Matulonis, Dana-Farber Cancer Institute; Michael J. Birrer, Massachusetts General Hospital, Boston; Rodrigo Ruiz-Soto, ImmunoGen, Waltham, MA; Jason A. Konner, Memorial Sloan Kettering Cancer Center, New York, NY; Raymond P. Perez, University of Kansas, Fairway, KS; and Todd M. Bauer, Sarah Cannon Research Institute and Tennessee Oncology, Nashville, TN
| | - Ursula A. Matulonis
- Kathleen N. Moore, University of Oklahoma Health Sciences Center, Oklahoma City, OK; Lainie P. Martin, Fox Chase Cancer Center, Philadelphia, PA; David M. O’Malley, Ohio State University James Cancer Center, Columbus, OH; Ursula A. Matulonis, Dana-Farber Cancer Institute; Michael J. Birrer, Massachusetts General Hospital, Boston; Rodrigo Ruiz-Soto, ImmunoGen, Waltham, MA; Jason A. Konner, Memorial Sloan Kettering Cancer Center, New York, NY; Raymond P. Perez, University of Kansas, Fairway, KS; and Todd M. Bauer, Sarah Cannon Research Institute and Tennessee Oncology, Nashville, TN
| | - Jason A. Konner
- Kathleen N. Moore, University of Oklahoma Health Sciences Center, Oklahoma City, OK; Lainie P. Martin, Fox Chase Cancer Center, Philadelphia, PA; David M. O’Malley, Ohio State University James Cancer Center, Columbus, OH; Ursula A. Matulonis, Dana-Farber Cancer Institute; Michael J. Birrer, Massachusetts General Hospital, Boston; Rodrigo Ruiz-Soto, ImmunoGen, Waltham, MA; Jason A. Konner, Memorial Sloan Kettering Cancer Center, New York, NY; Raymond P. Perez, University of Kansas, Fairway, KS; and Todd M. Bauer, Sarah Cannon Research Institute and Tennessee Oncology, Nashville, TN
| | - Raymond P. Perez
- Kathleen N. Moore, University of Oklahoma Health Sciences Center, Oklahoma City, OK; Lainie P. Martin, Fox Chase Cancer Center, Philadelphia, PA; David M. O’Malley, Ohio State University James Cancer Center, Columbus, OH; Ursula A. Matulonis, Dana-Farber Cancer Institute; Michael J. Birrer, Massachusetts General Hospital, Boston; Rodrigo Ruiz-Soto, ImmunoGen, Waltham, MA; Jason A. Konner, Memorial Sloan Kettering Cancer Center, New York, NY; Raymond P. Perez, University of Kansas, Fairway, KS; and Todd M. Bauer, Sarah Cannon Research Institute and Tennessee Oncology, Nashville, TN
| | - Todd M. Bauer
- Kathleen N. Moore, University of Oklahoma Health Sciences Center, Oklahoma City, OK; Lainie P. Martin, Fox Chase Cancer Center, Philadelphia, PA; David M. O’Malley, Ohio State University James Cancer Center, Columbus, OH; Ursula A. Matulonis, Dana-Farber Cancer Institute; Michael J. Birrer, Massachusetts General Hospital, Boston; Rodrigo Ruiz-Soto, ImmunoGen, Waltham, MA; Jason A. Konner, Memorial Sloan Kettering Cancer Center, New York, NY; Raymond P. Perez, University of Kansas, Fairway, KS; and Todd M. Bauer, Sarah Cannon Research Institute and Tennessee Oncology, Nashville, TN
| | - Rodrigo Ruiz-Soto
- Kathleen N. Moore, University of Oklahoma Health Sciences Center, Oklahoma City, OK; Lainie P. Martin, Fox Chase Cancer Center, Philadelphia, PA; David M. O’Malley, Ohio State University James Cancer Center, Columbus, OH; Ursula A. Matulonis, Dana-Farber Cancer Institute; Michael J. Birrer, Massachusetts General Hospital, Boston; Rodrigo Ruiz-Soto, ImmunoGen, Waltham, MA; Jason A. Konner, Memorial Sloan Kettering Cancer Center, New York, NY; Raymond P. Perez, University of Kansas, Fairway, KS; and Todd M. Bauer, Sarah Cannon Research Institute and Tennessee Oncology, Nashville, TN
| | - Michael J. Birrer
- Kathleen N. Moore, University of Oklahoma Health Sciences Center, Oklahoma City, OK; Lainie P. Martin, Fox Chase Cancer Center, Philadelphia, PA; David M. O’Malley, Ohio State University James Cancer Center, Columbus, OH; Ursula A. Matulonis, Dana-Farber Cancer Institute; Michael J. Birrer, Massachusetts General Hospital, Boston; Rodrigo Ruiz-Soto, ImmunoGen, Waltham, MA; Jason A. Konner, Memorial Sloan Kettering Cancer Center, New York, NY; Raymond P. Perez, University of Kansas, Fairway, KS; and Todd M. Bauer, Sarah Cannon Research Institute and Tennessee Oncology, Nashville, TN
| |
Collapse
|
194
|
Tolcher A. Antibody drug conjugates: lessons from 20 years of clinical experience. Ann Oncol 2016; 27:2168-2172. [DOI: 10.1093/annonc/mdw424] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/25/2016] [Accepted: 08/26/2016] [Indexed: 11/13/2022] Open
|
195
|
|
196
|
Donaghy H. Effects of antibody, drug and linker on the preclinical and clinical toxicities of antibody-drug conjugates. MAbs 2016; 8:659-71. [PMID: 27045800 PMCID: PMC4966843 DOI: 10.1080/19420862.2016.1156829] [Citation(s) in RCA: 343] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 02/15/2016] [Accepted: 02/16/2016] [Indexed: 11/15/2022] Open
Abstract
Antibody-drug conjugates (ADCs) represent a new class of cancer therapeutics. Their design involves a tumor-specific antibody, a linker and a cytotoxic payload. They were designed to allow specific targeting of highly potent cytotoxic agents to tumor cells whilst sparing normal cells. Frequent toxicities that may be driven by any of the components of an ADC have been reported. There are currently more than 50 ADCs in active clinical development, and a further ∼20 that have been discontinued. For this review, the reported toxicities of ADCs were analysed, and the mechanisms for their effects are explored in detail. Methods to reduce toxicities, including dosing strategies and drug design, are discussed. The toxicities reported for active and discontinued drugs are important to drive the rational design and improve the therapeutic index of ADCs of the future.
Collapse
|