1
|
He Y, Tian R, Xu D, Wu Y, Rina S, Chen T, Guan Y, Xie T, Ying T, Xie F, Han J. Preclinical evaluation and pilot clinical study of [ 68Ga]Ga-NOTA-H006 for non-invasive PET imaging of 5T4 oncofetal antigen. Eur J Nucl Med Mol Imaging 2025; 52:611-622. [PMID: 39377811 DOI: 10.1007/s00259-024-06941-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
PURPOSE Trophoblast glycoprotein, the so-called 5T4, is an oncofetal antigen expressed in many different cancers. However, no 5T4-specific radioligand is employed in the clinic for non-invasive diagnosis. Thus, the aim of the current study was to develop a PET radiotracer for imaging 5T4 expression in preclinical and clinical stages. METHODS A VHH library was constructed by camel immunization. The specificity of the VHHs toward 5T4 antigen was screened through phage display biopanning and periplasmic extract enzyme-linked immunosorbent assay. 1,4,7-Triazacyclononane-1,4,7-triacetate acid (NOTA) derivative was conjugated to the selected VHH. After radiolabeling, microPET/CT and ex vivo biodistribution were conducted using BxPC-3 and MDA-MB-468 tumor-bearing mice. Cold VHH was co-injected with the tracer to challenge its binding in vivo. For the pilot clinical study, PET/CT images were acquired at 1 h after injection of tracer in patients with pathologically confirmed primary and metastatic tumors. RESULTS A library with a capacity of 1.2 × 1012 colony-forming units was constructed after successful camel immunization. Nb1-40 with a median effect concentration of 0.43 nM was selected. After humanization, the resulting H006 maintained a high affinity towards 5T4. [68Ga]Ga-NOTA-H006 with the molar activities of 6.48-54.2 GBq/µmol was prepared with high radiochemical purity (> 98%). Using [68Ga]Ga-NOTA-H006, microPET/CT revealed a clear visualization of 5T4 expression in BxPC-3 tumor-bearing mice. Ex vivo biodistribution showed that the highest tumor-to-blood ratio (∼ 3-fold) and tumor-to-muscle ratio (∼ 5-fold) were achieved at 60 min post-injection. Co-injection of the cold H006 at a dose of 1.5 mg/kg significantly reduced the tumor uptake (p < 0.0001). In the pilot clinical study, [68Ga]Ga-NOTA-H006 demonstrated its capacity to map 5T4-positive lesions in humans and yielded a mean effective dose of 3.4 × 10- 2 mSv/MBq. CONCLUSIONS [68Ga]Ga-NOTA-H006, which can visualize 5T4 expression in vivo, has been successfully developed. This opens up opportunities for non-invasively studying 5T4 expression through nuclear medicine. Further clinical investigations are warranted to explore its clinical value in disease progression and companion diagnosis.
Collapse
Affiliation(s)
- Yingfang He
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai, 200032, China
| | - Ruhua Tian
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Dong Xu
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanfei Wu
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200233, China
| | - Sa Rina
- Huahe Pharmaceutical Co., Ltd, Shanghai, China
| | - Tengxiang Chen
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200233, China
| | - Tianwu Xie
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai, 200032, China
| | - Tianlei Ying
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200233, China.
| | - Junbin Han
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai, 200032, China.
| |
Collapse
|
2
|
Yang Y, Patel JM, Yang RS, Ma F, Niu X, Zhang Y, Niedringhaus T, Al-Sayah M, Yang X. Determination of the decapping efficiency of THIOMAB™ antibodies with the engineered cysteine in the Fc region for making antibody-drug conjugates by specific hinge fragmentation-liquid chromatography. Anal Bioanal Chem 2024:10.1007/s00216-024-05707-w. [PMID: 39688670 DOI: 10.1007/s00216-024-05707-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024]
Abstract
The site-specific antibody-drug conjugates (ADCs), particularly those utilizing the engineered cysteine in Fc fragments of mAbs (THIOMAB™ antibodies), have emerged as a novel class of biotherapeutics for cancer treatment. The engineered cysteine residues in these antibodies are capped by cysteine or glutathione through a disulfide bond. Prior to conjugation with linker-payloads, these caps need to be removed through a reduction process. However, monitoring the efficiency of the decapping process has been challenging due to the lack of effective analytical methods. Intact reversed-phase liquid chromatography-mass spectrometry and hydrophobic interaction chromatography methods failed to separate decapped and capped intact THIOMAB™ mAbs in our study. Instead the fragmentation of mAbs provided a novel strategy to analyze the decapping effiency. After cleavage using a hinge specific enzyme, the generated Fc fragments with and without cysteine and/or glutathione caps displayed different hydrophobicity and were well separated by RPLC, allowing quantitative determination of the decapping efficiency. Enzymes that cleave both above and below the hinge disulfide bonds were tested. The use of FabALATICA can determine percentages of molecules with 0, 1, and 2 cysteine and/or glutathione caps, respectively, regardless of whether the antibody contains the hinge LALA mutations. On the other hand, FabRICATOR enzyme can only be utilized for antibodies without LALA mutations for the overall decapping percentage and cannot be used to estimate intact antibody each with 0, 1, and 2 caps. Therefore, FabALACTICA cleavage followed by RPLC provides a wider application of monitoring the decapping efficiency of all antibodies with the engineered cysteine in Fc.
Collapse
Affiliation(s)
- Yun Yang
- Discovery Analytical Research, Merck & Co., Inc, 213 E Grand Ave, South San Francisco, CA, 94080, USA
| | - Jaymin M Patel
- Biologics Analytical Research and Development (BARD), Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, NJ, 07065, USA
| | - Rong-Sheng Yang
- Analytical Research and Development, Merck & Co., Inc, 126 E. Lincoln Avenue, Rahway, NJ, 07065, USA
| | - Fengfei Ma
- Discovery Analytical Research, Merck & Co., Inc, 213 E Grand Ave, South San Francisco, CA, 94080, USA
| | - Xiangfeng Niu
- Discovery Analytical Research, Merck & Co., Inc, 213 E Grand Ave, South San Francisco, CA, 94080, USA
| | - Yixiao Zhang
- Bioprocess Research & Development, Merck & Co., Inc, 126 E. Lincoln Avenue, Rahway, NJ, 07065, USA
| | - Thomas Niedringhaus
- Biologics Analytical Research and Development (BARD), Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, NJ, 07065, USA
| | - Mohammad Al-Sayah
- Discovery Analytical Research, Merck & Co., Inc, 213 E Grand Ave, South San Francisco, CA, 94080, USA
| | - Xiaoyu Yang
- Discovery Analytical Research, Merck & Co., Inc, 213 E Grand Ave, South San Francisco, CA, 94080, USA.
- Biologics Analytical Research and Development (BARD), Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, NJ, 07065, USA.
| |
Collapse
|
3
|
Adhikari P, Li G, Go M, Mandikian D, Rafidi H, Ng C, Anifa S, Johnson K, Bao L, Hernandez Barry H, Rowntree R, Agard N, Wu C, Chou KJ, Zhang D, Kozak KR, Pillow TH, Lewis GD, Yu SF, Boswell CA, Sadowsky JD. On Demand Bioorthogonal Switching of an Antibody-Conjugated SPECT Probe to a Cytotoxic Payload: from Imaging to Therapy. J Am Chem Soc 2024; 146:19088-19100. [PMID: 38946086 DOI: 10.1021/jacs.4c03529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Antibody-drug conjugates (ADCs) for the treatment of cancer aim to achieve selective delivery of a cytotoxic payload to tumor cells while sparing normal tissue. In vivo, multiple tumor-dependent and -independent processes act on ADCs and their released payloads to impact tumor-versus-normal delivery, often resulting in a poor therapeutic window. An ADC with a labeled payload would make synchronous correlations between distribution and tissue-specific pharmacological effects possible, empowering preclinical and clinical efforts to improve tumor-selective delivery; however, few methods to label small molecules without destroying their pharmacological activity exist. Herein, we present a bioorthogonal switch approach that allows a radiolabel attached to an ADC payload to be removed tracelessly at will. We exemplify this approach with a potent DNA-damaging agent, the pyrrolobenzodiazepine (PBD) dimer, delivered as an antibody conjugate targeted to lung tumor cells. The radiometal chelating group, DOTA, was attached via a novel trans-cyclooctene (TCO)-caged self-immolative para-aminobenzyl (PAB) linker to the PBD, stably attenuating payload activity and allowing tracking of biodistribution in tumor-bearing mice via SPECT-CT imaging (live) or gamma counting (post-mortem). Following TCO-PAB-DOTA reaction with tetrazines optimized for extra- and intracellular reactivity, the label was removed to reveal the unmodified PBD dimer capable of inducing potent tumor cell killing in vitro and in mouse xenografts. The switchable antibody radio-drug conjugate (ArDC) we describe integrates, but decouples, the two functions of a theranostic given that it can serve as a diagnostic for payload delivery in the labeled state, but can be switched on demand to a therapeutic agent (an ADC).
Collapse
Affiliation(s)
- Pragya Adhikari
- Genentech Inc., South San Francisco, California 94080, United States
| | - Guangmin Li
- Genentech Inc., South San Francisco, California 94080, United States
| | - MaryAnn Go
- Genentech Inc., South San Francisco, California 94080, United States
| | | | - Hanine Rafidi
- Genentech Inc., South San Francisco, California 94080, United States
| | - Carl Ng
- Genentech Inc., South San Francisco, California 94080, United States
| | - Sagana Anifa
- Genentech Inc., South San Francisco, California 94080, United States
| | - Kevin Johnson
- Genentech Inc., South San Francisco, California 94080, United States
| | - Linda Bao
- Genentech Inc., South San Francisco, California 94080, United States
| | | | - Rebecca Rowntree
- Genentech Inc., South San Francisco, California 94080, United States
| | - Nicholas Agard
- Genentech Inc., South San Francisco, California 94080, United States
| | - Cong Wu
- Genentech Inc., South San Francisco, California 94080, United States
| | - Kang-Jye Chou
- Genentech Inc., South San Francisco, California 94080, United States
| | - Donglu Zhang
- Genentech Inc., South San Francisco, California 94080, United States
| | - Katherine R Kozak
- Genentech Inc., South San Francisco, California 94080, United States
| | - Thomas H Pillow
- Genentech Inc., South San Francisco, California 94080, United States
| | - Gail D Lewis
- Genentech Inc., South San Francisco, California 94080, United States
| | - Shang-Fan Yu
- Genentech Inc., South San Francisco, California 94080, United States
| | - C Andrew Boswell
- Genentech Inc., South San Francisco, California 94080, United States
| | - Jack D Sadowsky
- Genentech Inc., South San Francisco, California 94080, United States
| |
Collapse
|
4
|
Khadela A, Megha K, Shah VB, Soni S, Shah AC, Mistry H, Bhatt S, Merja M. Exploring the Potential of Antibody-Drug Conjugates in Targeting Non-small Cell Lung Cancer Biomarkers. Clin Med Insights Oncol 2024; 18:11795549241260534. [PMID: 38911453 PMCID: PMC11193349 DOI: 10.1177/11795549241260534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 05/17/2024] [Indexed: 06/25/2024] Open
Abstract
Antibody-drug conjugates (ADCs), combining the cytotoxicity of the drug payload with the specificity of monoclonal antibodies, are one of the rapidly evolving classes of anti-cancer agents. These agents have been successfully incorporated into the treatment paradigm of many malignancies, including non-small cell lung cancer (NSCLC). The NSCLC is the most prevalent subtype of lung cancer, having a considerable burden on the cancer-related mortality and morbidity rates globally. Several ADC molecules are currently approved by the Food and Drug Administration (FDA) to be used in patients with NSCLC. However, the successful management of NSCLC patients using these agents was met with several challenges, including the development of resistance and toxicities. These shortcomings resulted in the exploration of novel therapeutic targets that can be targeted by the ADCs. This review aims to explore the recently identified ADC targets along with their oncologic mechanisms. The ADC molecules targeting these biomarkers are further discussed along with the evidence from clinical trials.
Collapse
Affiliation(s)
- Avinash Khadela
- Department of Pharmacology, L. M. College of Pharmacy, Navrangpura, Ahmedabad, Gujarat, India
| | - Kaivalya Megha
- Department of Pharmacology, L. M. College of Pharmacy, Navrangpura, Ahmedabad, Gujarat, India
| | - Vraj B Shah
- Department of Pharmacology, L. M. College of Pharmacy, Navrangpura, Ahmedabad, Gujarat, India
| | - Shruti Soni
- Department of Pharmacology, L. M. College of Pharmacy, Navrangpura, Ahmedabad, Gujarat, India
| | - Aayushi C Shah
- Department of Pharmacology, L. M. College of Pharmacy, Navrangpura, Ahmedabad, Gujarat, India
| | - Hetvi Mistry
- Department of Pharmacology, L. M. College of Pharmacy, Navrangpura, Ahmedabad, Gujarat, India
| | - Shelly Bhatt
- Department of Pharmacology, L. M. College of Pharmacy, Navrangpura, Ahmedabad, Gujarat, India
| | - Manthan Merja
- Department of Clinical Oncology, Starlit Cancer Centre, Kothiya Hospital, Ahmedabad, Gujarat, India
| |
Collapse
|
5
|
Nelson MH, Fritzell S, Miller R, Werchau D, Van Citters D, Nilsson A, Misher L, Ljung L, Bader R, Deronic A, Chunyk AG, Schultz L, Varas LA, Rose N, Håkansson M, Gross J, Furebring C, Pavlik P, Sundstedt A, Veitonmäki N, Ramos HJ, Säll A, Dahlman A, Bienvenue D, von Schantz L, McMahan CJ, Askmyr M, Hernandez-Hoyos G, Ellmark P. The Bispecific Tumor Antigen-Conditional 4-1BB x 5T4 Agonist, ALG.APV-527, Mediates Strong T-Cell Activation and Potent Antitumor Activity in Preclinical Studies. Mol Cancer Ther 2023; 22:89-101. [PMID: 36343381 PMCID: PMC9808321 DOI: 10.1158/1535-7163.mct-22-0395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/16/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022]
Abstract
4-1BB (CD137) is an activation-induced costimulatory receptor that regulates immune responses of activated CD8 T and natural killer cells, by enhancing proliferation, survival, cytolytic activity, and IFNγ production. The ability to induce potent antitumor activity by stimulating 4-1BB on tumor-specific cytotoxic T cells makes 4-1BB an attractive target for designing novel immuno-oncology therapeutics. To minimize systemic immune toxicities and enhance activity at the tumor site, we have developed a novel bispecific antibody that stimulates 4-1BB function when co-engaged with the tumor-associated antigen 5T4. ALG.APV-527 was built on the basis of the ADAPTIR bispecific platform with optimized binding domains to 4-1BB and 5T4 originating from the ALLIGATOR-GOLD human single-chain variable fragment library. The epitope of ALG.APV-527 was determined to be located at domain 1 and 2 on 4-1BB using X-ray crystallography. As shown in reporter and primary cell assays in vitro, ALG.APV-527 triggers dose-dependent 4-1BB activity mediated only by 5T4 crosslinking. In vivo, ALG.APV-527 demonstrates robust antitumor responses, by inhibiting growth of established tumors expressing human 5T4 followed by a long-lasting memory immune response. ALG.APV-527 has an antibody-like half-life in cynomolgus macaques and was well tolerated at 50.5 mg/kg. ALG.APV-527 is uniquely designed for 5T4-conditional 4-1BB-mediated antitumor activity with potential to minimize systemic immune activation and hepatotoxicity while providing efficacious tumor-specific responses in a range of 5T4-expressing tumor indications as shown by robust activity in preclinical in vitro and in vivo models. On the basis of the combined preclinical dataset, ALG.APV-527 has potential as a promising anticancer therapeutic for the treatment of 5T4-expressing tumors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Jane Gross
- Aptevo Therapeutics Inc., Seattle, Washington
| | | | | | | | | | | | - Anna Säll
- Alligator Bioscience AB, Lund, Sweden
| | | | | | | | | | | | | | - Peter Ellmark
- Alligator Bioscience AB, Lund, Sweden.,Department of Immunotechnology, Lund University, Lund, Sweden.,Corresponding Author: Peter Ellmark, Alligator Bioscience, Medicon Village, 223 81 Lund, Sweden. Phone: 467-9721-2739; E-mail:
| |
Collapse
|
6
|
Kemper K, Gielen E, Boross P, Houtkamp M, Plantinga TS, de Poot SAH, Burm SM, Janmaat ML, Koopman LA, van den Brink EN, Rademaker R, Verzijl D, Engelberts PJ, Satijn D, Sasser AK, Breij ECW. Mechanistic and pharmacodynamic studies of DuoBody-CD3x5T4 in preclinical tumor models. Life Sci Alliance 2022; 5:5/11/e202201481. [PMID: 36271507 PMCID: PMC9458754 DOI: 10.26508/lsa.202201481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/24/2022] Open
Abstract
CD3 bispecific antibodies (bsAbs) show great promise as anticancer therapeutics. Here, we show in-depth mechanistic studies of a CD3 bsAb in solid cancer, using DuoBody-CD3x5T4. Cross-linking T cells with tumor cells expressing the oncofetal antigen 5T4 was required to induce cytotoxicity. Naive and memory CD4+ and CD8+ T cells were equally effective at mediating cytotoxicity, and DuoBody-CD3x5T4 induced partial differentiation of naive T-cell subsets into memory-like cells. Tumor cell kill was associated with T-cell activation, proliferation, and production of cytokines, granzyme B, and perforin. Genetic knockout of FAS or IFNGR1 in 5T4+ tumor cells abrogated tumor cell kill. In the presence of 5T4+ tumor cells, bystander kill of 5T4− but not of 5T4−IFNGR1− tumor cells was observed. In humanized xenograft models, DuoBody-CD3x5T4 antitumor activity was associated with intratumoral and peripheral blood T-cell activation. Lastly, in dissociated patient-derived tumor samples, DuoBody-CD3x5T4 activated tumor-infiltrating lymphocytes and induced tumor-cell cytotoxicity, even when most tumor-infiltrating lymphocytes expressed PD-1. These data provide an in-depth view on the mechanism of action of a CD3 bsAb in preclinical models of solid cancer.
Collapse
|
7
|
Discovery of Novel Polyamide-Pyrrolobenzodiazepine Hybrids for Antibody-Drug Conjugates. Bioorg Med Chem Lett 2022; 72:128876. [PMID: 35788036 DOI: 10.1016/j.bmcl.2022.128876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/31/2022] [Accepted: 06/27/2022] [Indexed: 11/21/2022]
Abstract
Pyrrolobenzodiazepine (PBD) dimers are well-known highly potent antibody drug conjugate (ADC) payloads. The corresponding PBD monomers, in contrast, have received much less attention from the ADC community. We prepared several novel polyamide-linked PBD monomers and evaluated their utility as ADC payloads. The unconjugated polyamide-PBD hybrids exhibited potent antiproliferative activity (IC50 range: 10-11-10-8 M) against a variety of HER2-expressing cancer cell lines. Several peptide-linked variants of the lead compound were prepared and conjugated to trastuzumab to afford ADCs with drug-to-antibody (DAR) ratios ranging from 3-5. The ADCs exhibited antigen-dependent cytotoxicity in vitro and potently suppressed tumor xenograft growth in vivo in a target-dependent manner. Moreover, the ADCs were well-tolerated in both mouse and rat. This work demonstrates for the first time that PBD polyamide hybrids can serve as effective ADC payloads.
Collapse
|
8
|
Teicher BA, Morris J. Antibody-Drug Conjugate Targets, Drugs and Linkers. Curr Cancer Drug Targets 2022; 22:463-529. [PMID: 35209819 DOI: 10.2174/1568009622666220224110538] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/22/2021] [Accepted: 11/09/2021] [Indexed: 11/22/2022]
Abstract
Antibody-drug conjugates offer the possibility of directing powerful cytotoxic agents to a malignant tumor while sparing normal tissue. The challenge is to select an antibody target expressed exclusively or at highly elevated levels on the surface of tumor cells and either not all or at low levels on normal cells. The current review explores 78 targets that have been explored as antibody-drug conjugate targets. Some of these targets have been abandoned, 9 or more are the targets of FDA-approved drugs, and most remain active clinical interest. Antibody-drug conjugates require potent cytotoxic drug payloads, several of these small molecules are discussed, as are the linkers between the protein component and small molecule components of the conjugates. Finally, conclusions regarding the elements for the successful antibody-drug conjugate are discussed.
Collapse
Affiliation(s)
- Beverly A Teicher
- Developmental Therapeutics Program, DCTD, National Cancer Institute, Bethesda, MD 20892,United States
| | - Joel Morris
- Developmental Therapeutics Program, DCTD, National Cancer Institute, Bethesda, MD 20892,United States
| |
Collapse
|
9
|
Smith RA, Zammit DJ, Damle NK, Usansky H, Reddy SP, Lin JH, Mistry M, Rao NS, Denis LJ, Gupta S. ASN004, A 5T4-targeting scFv-Fc Antibody-Drug Conjugate with High Drug-to-Antibody Ratio, Induces Complete and Durable Tumor Regressions in Preclinical Models. Mol Cancer Ther 2021; 20:1327-1337. [PMID: 34045226 DOI: 10.1158/1535-7163.mct-20-0565] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 03/24/2021] [Accepted: 05/10/2021] [Indexed: 11/16/2022]
Abstract
The 5T4 oncofetal antigen (trophoblast glycoprotein) is expressed in a wide range of malignant tumors but shows very limited expression in normal adult tissues. ASN004 is a 5T4-targeted antibody-drug conjugate (ADC) that incorporates a novel single-chain Fv-Fc antibody and Dolaflexin drug-linker technology, with an Auristatin F hydroxypropylamide payload drug-to-antibody ratio of approximately 10-12. The pharmacology, toxicology, and pharmacokinetic properties of ASN004 and its components were investigated in vitro and in vivo ASN004 showed high affinity for the 5T4 antigen and was selectively bound to and internalized into 5T4-expressing tumor cells, and potent cytotoxicity was demonstrated for a diverse panel of solid tumor cell lines. ASN004 induced complete and durable tumor regression in multiple tumor xenograft models, derived from human lung, breast, cervical, and gastric tumor cell lines having a wide range of 5T4 expression levels. A single dose of ASN004, as low as 1 mg/kg i.v., achieved complete tumor regression leading to tumor-free survivors in the A431 cervical cancer model. In head-to-head studies, superior activity of ASN004 was demonstrated against trastuzumab-DM1, in a low-5T4/high-HER2 expressing gastric tumor model, and 10-fold greater potency was found for ASN004 against the 5T4-targeted ADC PF-06263507 in a lung tumor model. In marmoset monkeys, ASN004 was well tolerated at doses up to 1.5 mg/kg Q3W i.v., and showed dose-dependent exposure, linear pharmacokinetics, and markedly low exposure of free payload drug. Taken together, these findings identify ASN004 as a promising new ADC therapeutic for clinical evaluation in a broad range of solid tumor types.
Collapse
Affiliation(s)
- Roger A Smith
- Asana BioSciences, Princeton Pike Corporate Center, Lawrenceville, New Jersey.
| | - David J Zammit
- Asana BioSciences, Princeton Pike Corporate Center, Lawrenceville, New Jersey
| | - Nitin K Damle
- Asana BioSciences, Princeton Pike Corporate Center, Lawrenceville, New Jersey
| | - Helen Usansky
- Asana BioSciences, Princeton Pike Corporate Center, Lawrenceville, New Jersey
| | - Sanjeeva P Reddy
- Asana BioSciences, Princeton Pike Corporate Center, Lawrenceville, New Jersey
| | - Jun-Hsiang Lin
- Asana BioSciences, Princeton Pike Corporate Center, Lawrenceville, New Jersey
| | - Mahesh Mistry
- Asana BioSciences, Princeton Pike Corporate Center, Lawrenceville, New Jersey
| | - Niranjan S Rao
- Asana BioSciences, Princeton Pike Corporate Center, Lawrenceville, New Jersey
| | - Louis J Denis
- Asana BioSciences, Princeton Pike Corporate Center, Lawrenceville, New Jersey
| | - Sandeep Gupta
- Asana BioSciences, Princeton Pike Corporate Center, Lawrenceville, New Jersey
| |
Collapse
|
10
|
Exatecan Antibody Drug Conjugates Based on a Hydrophilic Polysarcosine Drug-Linker Platform. Pharmaceuticals (Basel) 2021; 14:ph14030247. [PMID: 33803327 PMCID: PMC8000490 DOI: 10.3390/ph14030247] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/02/2021] [Accepted: 03/06/2021] [Indexed: 12/20/2022] Open
Abstract
We herein report the development and evaluation of a novel HER2-targeting antibody-drug conjugate (ADC) based on the topoisomerase I inhibitor payload exatecan, using our hydrophilic monodisperse polysarcosine (PSAR) drug-linker platform (PSARlink). In vitro and in vivo experiments were conducted in breast and gastric cancer models to characterize this original ADC and gain insight about the drug-linker structure-activity relationship. The inclusion of the PSAR hydrophobicity masking entity efficiently reduced the overall hydrophobicity of the conjugate and yielded an ADC sharing the same pharmacokinetic profile as the unconjugated antibody despite the high drug-load of the camptothecin-derived payload (drug-antibody ratio of 8). Tra-Exa-PSAR10 demonstrated strong anti-tumor activity at 1 mg/kg in an NCI-N87 xenograft model, outperforming the FDA-approved ADC DS-8201a (Enhertu), while being well tolerated in mice at a dose of 100 mg/kg. In vitro experiments showed that this exatecan-based ADC demonstrated higher bystander killing effect than DS-8201a and overcame resistance to T-DM1 (Kadcyla) in preclinical HER2+ breast and esophageal models, suggesting potential activity in heterogeneous and resistant tumors. In summary, the polysarcosine-based hydrophobicity masking approach allowsfor the generation of highly conjugated exatecan-based ADCs having excellent physicochemical properties, an improved pharmacokinetic profile, and potent in vivo anti-tumor activity.
Collapse
|
11
|
Mao S, Chaerkady R, Yu W, D'Angelo G, Garcia A, Chen H, Barrett AM, Phipps S, Fleming R, Hess S, Koopmann JO, Dimasi N, Wilson S, Pugh K, Cook K, Masterson LA, Gao C, Wu H, Herbst R, Howard PW, Tice DA, Cobbold M, Harper J. Resistance to Pyrrolobenzodiazepine Dimers Is Associated with SLFN11 Downregulation and Can Be Reversed through Inhibition of ATR. Mol Cancer Ther 2021; 20:541-552. [PMID: 33653945 DOI: 10.1158/1535-7163.mct-20-0351] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 11/01/2020] [Accepted: 01/07/2021] [Indexed: 11/16/2022]
Abstract
Resistance to antibody-drug conjugates (ADCs) has been observed in both preclinical models and clinical studies. However, mechanisms of resistance to pyrrolobenzodiazepine (PBD)-conjugated ADCs have not been well characterized and thus, this study was designed to investigate development of resistance to PBD dimer warheads and PBD-conjugated ADCs. We established a PBD-resistant cell line, 361-PBDr, by treating human breast cancer MDA-MB-361 cells with gradually increasing concentrations of SG3199, the PBD dimer released from the PBD drug-linker tesirine. 361-PBDr cells were over 20-fold less sensitive to SG3199 compared with parental cells and were cross-resistant to other PBD warhead and ADCs conjugated with PBDs. Proteomic profiling revealed that downregulation of Schlafen family member 11 (SLFN11), a putative DNA/RNA helicase, sensitizing cancer cells to DNA-damaging agents, was associated with PBD resistance. Confirmatory studies demonstrated that siRNA knockdown of SLFN11 in multiple tumor cell lines conferred reduced sensitivity to SG3199 and PBD-conjugated ADCs. Treatment with EPZ011989, an EZH2 inhibitor, derepressed SLFN11 expression in 361-PBDr and other SLFN11-deficient tumor cells, and increased sensitivity to PBD and PBD-conjugated ADCs, indicating that the suppression of SLFN11 expression is associated with histone methylation as reported. Moreover, we demonstrated that combining an ataxia telangiectasia and Rad3-related protein (ATR) inhibitor, AZD6738, with SG3199 or PBD-based ADCs led to synergistic cytotoxicity in either resistant 361-PBDr cells or cells that SLFN11 was knocked down via siRNA. Collectively, these data provide insights into potential development of resistance to PBDs and PBD-conjugated ADCs, and more importantly, inform strategy development to overcome such resistance.
Collapse
Affiliation(s)
- Shenlan Mao
- Early Oncology R&D, AstraZeneca, Gaithersburg, Maryland.
| | | | - Wen Yu
- Bioinformatics, AstraZeneca, Gaithersburg, Maryland
| | | | - Andrew Garcia
- Antibody Discovery & Protein Engineering, AstraZeneca, Gaithersburg, Maryland
| | - Hong Chen
- Early Oncology R&D, AstraZeneca, Gaithersburg, Maryland
| | | | - Sandrina Phipps
- Antibody Discovery & Protein Engineering, AstraZeneca, Gaithersburg, Maryland
| | - Ryan Fleming
- Antibody Discovery & Protein Engineering, AstraZeneca, Gaithersburg, Maryland
| | - Sonja Hess
- Antibody Discovery & Protein Engineering, AstraZeneca, Gaithersburg, Maryland
| | | | - Nazzareno Dimasi
- Antibody Discovery & Protein Engineering, AstraZeneca, Gaithersburg, Maryland
| | - Susan Wilson
- Antibody Discovery & Protein Engineering, AstraZeneca, Gaithersburg, Maryland
| | | | - Kimberly Cook
- Early Oncology R&D, AstraZeneca, Gaithersburg, Maryland
| | | | - Changshou Gao
- Antibody Discovery & Protein Engineering, AstraZeneca, Gaithersburg, Maryland
| | - Herren Wu
- Antibody Discovery & Protein Engineering, AstraZeneca, Gaithersburg, Maryland
| | - Ronald Herbst
- Early Oncology R&D, AstraZeneca, Gaithersburg, Maryland
| | | | - David A Tice
- Early Oncology R&D, AstraZeneca, Gaithersburg, Maryland
| | - Mark Cobbold
- Early Oncology R&D, AstraZeneca, Gaithersburg, Maryland
| | - Jay Harper
- Early Oncology R&D, AstraZeneca, Gaithersburg, Maryland.
| |
Collapse
|
12
|
Walsh SJ, Bargh JD, Dannheim FM, Hanby AR, Seki H, Counsell AJ, Ou X, Fowler E, Ashman N, Takada Y, Isidro-Llobet A, Parker JS, Carroll JS, Spring DR. Site-selective modification strategies in antibody-drug conjugates. Chem Soc Rev 2021; 50:1305-1353. [PMID: 33290462 DOI: 10.1039/d0cs00310g] [Citation(s) in RCA: 237] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Antibody-drug conjugates (ADCs) harness the highly specific targeting capabilities of an antibody to deliver a cytotoxic payload to specific cell types. They have garnered widespread interest in drug discovery, particularly in oncology, as discrimination between healthy and malignant tissues or cells can be achieved. Nine ADCs have received approval from the US Food and Drug Administration and more than 80 others are currently undergoing clinical investigations for a range of solid tumours and haematological malignancies. Extensive research over the past decade has highlighted the critical nature of the linkage strategy adopted to attach the payload to the antibody. Whilst early generation ADCs were primarily synthesised as heterogeneous mixtures, these were found to have sub-optimal pharmacokinetics, stability, tolerability and/or efficacy. Efforts have now shifted towards generating homogeneous constructs with precise drug loading and predetermined, controlled sites of attachment. Homogeneous ADCs have repeatedly demonstrated superior overall pharmacological profiles compared to their heterogeneous counterparts. A wide range of methods have been developed in the pursuit of homogeneity, comprising chemical or enzymatic methods or a combination thereof to afford precise modification of specific amino acid or sugar residues. In this review, we discuss advances in chemical and enzymatic methods for site-specific antibody modification that result in the generation of homogeneous ADCs.
Collapse
Affiliation(s)
- Stephen J Walsh
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Richter J, Thibaud S. Anti-body building: The exercise of advancing immune based myeloma therapies. Blood Rev 2020; 48:100789. [PMID: 33384171 DOI: 10.1016/j.blre.2020.100789] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/27/2020] [Accepted: 12/16/2020] [Indexed: 12/26/2022]
Abstract
The last decade has seen a marked improvement in the outcomes of patients with multiple myeloma. Much of this has been due to not only the advent of new therapies, but their inherent ability to be combined into 3 and 4 drug regimens without resulting in unacceptable toxicity. The general gestalt has been to combine agents of varied mechanisms of action. With the primary classes of agents such as proteasome inhibitors and immunomodulatory drugs as bases, the advent of antibody-based therapy in myeloma has allowed us to easily augment these therapies; much in the same way rituximab impacted the lymphoma world. With the approvals of daratumumab, elotuzumab and isatuximab; the myeloma world was ushered into the next wave of targeted agents. Here, we take a look at the current landscape of "off-the-shelf" antibody-based therapies in myeloma and peer into the next wave of multi-functional targeted agents.
Collapse
Affiliation(s)
- Joshua Richter
- Tisch Cancer Institute: Icahn School of Medicine at Mount Sinai, USA.
| | - Santiago Thibaud
- Tisch Cancer Institute: Icahn School of Medicine at Mount Sinai, USA.
| |
Collapse
|
14
|
Hartley JA. Antibody-drug conjugates (ADCs) delivering pyrrolobenzodiazepine (PBD) dimers for cancer therapy. Expert Opin Biol Ther 2020; 21:931-943. [PMID: 32543981 DOI: 10.1080/14712598.2020.1776255] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
INTRODUCTION The rationally designed pyrrolobenzodiazepine (PBD) dimers emerged around ten years ago as a new class of drug component for antibody-drug conjugates (ADC). They produce highly cytotoxic DNA cross-links, exploiting a completely different cellular target to the auristatin and maytansinoid tubulin inhibitor classes and a different mode of DNA damage to other DNA interacting warheads such as calicheamicin. AREAS COVERED The properties which make the PBD dimers suitable warheads for ADCs, and the development of the two main payload structures talirine and tesirine, are discussed. The clinical experience with the twenty PBD dimer-containing ADCs to enter the clinic is reviewed, with a focus on vadastuximab talirine and rovalpituzumab tesirine, both of which were discontinued following pivotal studies, and loncastuximab tesirine and camidanlumab tesirine which are progressing towards approval. EXPERT OPINION Reviewing the clinical efficacy and safety data from almost forty clinical trials of PBD dimer-containing ADCs highlights the complexities and challenges of ADC early clinical development. It enables some conclusions to be made about reasons for failure and suggests strategies to optimise the future clinical development of this promising class of ADCs in a rapidly expanding field.
Collapse
Affiliation(s)
- John A Hartley
- Professor of Cancer Studies, UCL Cancer Institute, London, UK
| |
Collapse
|
15
|
Chen R, Rajan S, Overstreet MG, Hurt EM, Thomas SB, Muniz-Medina V, Ward C, Sadowska A, Fleming R, Karanth S, Breen S, Zheng B, Wu Y, Iverson WO, Novick S, O'Day T, Shah DP, Dimasi N, Tiberghien AC, Osbourn J, Walker J. Preclinical Characterization of an Antibody-Drug Conjugate Targeting CS-1 and the Identification of Uncharacterized Populations of CS-1-Positive Cells. Mol Cancer Ther 2020; 19:1649-1659. [PMID: 32404408 DOI: 10.1158/1535-7163.mct-19-0482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 11/21/2019] [Accepted: 05/08/2020] [Indexed: 11/16/2022]
Abstract
Multiple myeloma is a hematologic cancer that disrupts normal bone marrow function and has multiple lines of therapeutic options, but is incurable as patients ultimately relapse. We developed a novel antibody-drug conjugate (ADC) targeting CS-1, a protein that is highly expressed on multiple myeloma tumor cells. The anti-CS-1 mAb specifically bound to cells expressing CS-1 and, when conjugated to a cytotoxic pyrrolobenzodiazepine payload, reduced the viability of multiple myeloma cell lines in vitro In mouse models of multiple myeloma, a single administration of the CS-1 ADC caused durable regressions in disseminated models and complete regression in a subcutaneous model. In an exploratory study in cynomolgus monkeys, the CS-1 ADC demonstrated a half-life of 3 to 6 days; however, no highest nonseverely toxic dose was achieved, as bone marrow toxicity was dose limiting. Bone marrow from dosed monkeys showed reductions in progenitor cells as compared with normal marrow. In vitro cell killing assays demonstrated that the CS-1 ADC substantially reduced the number of progenitor cells in healthy bone marrow, leading us to identify previously unreported CS-1 expression on a small population of progenitor cells in the myeloid-erythroid lineage. This finding suggests that bone marrow toxicity is the result of both on-target and off-target killing by the ADC.
Collapse
Affiliation(s)
- Ruoyan Chen
- Research and Development, AstraZeneca, Gaithersburg, Maryland
| | - Saravanan Rajan
- Research and Development, AstraZeneca, Gaithersburg, Maryland
| | | | - Elaine M Hurt
- Research and Development, AstraZeneca, Gaithersburg, Maryland
| | | | | | | | | | - Ryan Fleming
- Research and Development, AstraZeneca, Gaithersburg, Maryland
| | | | - Shannon Breen
- Research and Development, AstraZeneca, Gaithersburg, Maryland
| | - Bo Zheng
- Research and Development, AstraZeneca, Gaithersburg, Maryland
| | - Yuling Wu
- Research and Development, AstraZeneca, Gaithersburg, Maryland
| | | | - Steven Novick
- Research and Development, AstraZeneca, Gaithersburg, Maryland
| | - Terrence O'Day
- Research and Development, AstraZeneca, Gaithersburg, Maryland
| | - Dipesha P Shah
- Research and Development, AstraZeneca, Gaithersburg, Maryland
| | | | | | - Jane Osbourn
- Research and Development, AstraZeneca, Cambridge, United Kingdom
| | - Jill Walker
- Research and Development, AstraZeneca, Gaithersburg, Maryland
| |
Collapse
|
16
|
Wan YL, Sapra P, Bolton J, Chua JX, Durrant LG, Stern PL. Combination Treatment with an Antibody-Drug Conjugate (A1mcMMAF) Targeting the Oncofetal Glycoprotein 5T4 and Carboplatin Improves Survival in a Xenograft Model of Ovarian Cancer. Target Oncol 2020; 14:465-477. [PMID: 31332693 PMCID: PMC6684567 DOI: 10.1007/s11523-019-00650-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Recurrence occurs in over 75% of women with epithelial ovarian cancer despite optimal treatment. Selectively killing tumour cells thought to initiate relapse using an antibody–drug conjugate could prolong progression-free survival and offer an improved side-effect profile. A1mcMMAF is an antibody–drug conjugate designed to target cells expressing the tumour-associated antigen 5T4. It has shown to be efficacious in various cell line models and have a greater impact when combined with routine chemotherapeutic regimes. Objectives This study aims to explore the potential for the use of a 5T4 antibody–drug conjugate in women with ovarian cancer both as a monotherapy and in combination with platinum-based chemotherapy. Methods Immunohistochemical analysis was used to assess 5T4 expression in tumours from patients with ovarian cancer. Effectiveness of A1mcMMAF therapy as a single agent and in combination with carboplatin was assessed in vitro in the ovarian cancer cell line SKOV3 and confirmed in vivo using a serial bioluminescence assay in a SKOV3 xenograft model of ovarian cancer. Results 5T4 is confirmed as suitably expressed in epithelial ovarian cancers prior to adjuvant therapy and is an independent predictor of poor survival. A1mcMMAF showed specific activity, both in vitro and in vivo, against SKOV3 ovarian cancer cells. When used in combination with carboplatin, in vivo tumour growth was inhibited resulting in prolonged survival in a SKOV3 xenograft model. Conclusions These data support further investigation of A1mcMMAF in combination with platinum-based chemotherapy in ovarian and other cancer treatments. Electronic supplementary material The online version of this article (10.1007/s11523-019-00650-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Y Louise Wan
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, 5th Floor Research, St Mary's Hospital, Oxford Road, Manchester, M13 9WL, UK
| | - Puja Sapra
- Oncology Research and Development, Pfizer Inc., 401 N. Middletown Road, Pearl River, NY, 10954, USA
| | - James Bolton
- Department of Histopathology, Manchester University NHS Foundation Trust, Oxford Road, Manchester, M13 9WL, UK
| | - Jia Xin Chua
- Academic Clinical Oncology, The University of Nottingham, City Hospital Campus, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Lindy G Durrant
- Academic Clinical Oncology, The University of Nottingham, City Hospital Campus, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Peter L Stern
- Manchester Cancer Research Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Wilmslow Road, Manchester, M20 4BX, UK.
| |
Collapse
|
17
|
Amani N, Dorkoosh FA, Mobedi H. ADCs, as Novel Revolutionary Weapons for Providing a Step Forward in Targeted Therapy of Malignancies. Curr Drug Deliv 2020; 17:23-51. [DOI: 10.2174/1567201816666191121145109] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 08/01/2019] [Accepted: 10/29/2019] [Indexed: 11/22/2022]
Abstract
:Antibody drug conjugates (ADCs), as potent pharmaceutical trojan horses for cancer treatment, provide superior efficacy and specific targeting along with low risk of adverse reactions compared to traditional chemotherapeutics. In fact, the development of these agents combines the selective targeting capability of monoclonal antibody (mAb) with high cytotoxicity of chemotherapeutics for controlling the neoplastic mass growth. Different ADCs (more than 60 ADCs) in preclinical and clinical trials were introduced in this novel pharmaceutical field. Various design-based factors must be taken into account for improving the functionality of ADC technology, including selection of appropriate target antigen and high binding affinity of fragment (miniaturized ADCs) or full mAbs (preferentially use of humanized or fully human antibodies compared to murine and chimeric ones), use of bispecific antibodies for dual targeting effect, linker engineering and conjugation method efficacy to obtain more controlled drug to antibody ratio (DAR). Challenging issues affecting therapeutic efficacy and safety of ADCs, including bystander effect, on- and off-target toxicities, multi drug resistance (MDR) are also addressed. 4 FDA-approved ADCs in the market, including ADCETRIS ®, MYLOTARG®, BESPONSA ®, KADCYLA®. The goal of the current review is to evaluate the key parameters affecting ADCs development.
Collapse
Affiliation(s)
- Nooshafarin Amani
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Abedin Dorkoosh
- Medical Biomaterial Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Mobedi
- Novel Drug Delivery Systems (NDDS) Department, Iran Polymer and Petrochemical Institute, Tehran, Iran
| |
Collapse
|
18
|
Campbell AD, Tomasi S, Tiberghien AC, Parker JS. An Isomerization Approach to Tesirine and Pyrrolobenzodiazepines. Org Process Res Dev 2019. [DOI: 10.1021/acs.oprd.9b00332] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Andrew D. Campbell
- Chemical Development, Pharmaceutical Technology & Development, AstraZeneca, Macclesfield SK10 2NA, United Kingdom
| | - Simone Tomasi
- Chemical Development, Pharmaceutical Technology & Development, AstraZeneca, Macclesfield SK10 2NA, United Kingdom
| | | | - Jeremy S. Parker
- Early Chemical Development, Pharmaceutical Sciences, R&D, AstraZeneca, Macclesfield, United Kingdom
| |
Collapse
|
19
|
Synthesis and evaluation of pyrrolobenzodiazepine dimer antibody-drug conjugates with dual β-glucuronide and dipeptide triggers. Eur J Med Chem 2019; 179:591-607. [DOI: 10.1016/j.ejmech.2019.06.044] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/07/2019] [Accepted: 06/16/2019] [Indexed: 12/17/2022]
|
20
|
Gregson SJ, Tiberghien AC, Masterson LA, Howard PW. Pyrrolobenzodiazepine Dimers as Antibody–Drug Conjugate (ADC) Payloads. CYTOTOXIC PAYLOADS FOR ANTIBODY – DRUG CONJUGATES 2019. [DOI: 10.1039/9781788012898-00296] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The pyrrolobenzodiazepine (PBD) ring system was first discovered in the 1960s and is found in several naturally occurring potent anti-tumour antibiotics. The mode of action of PBDs involves sequence-selective [purine–guanine–purine (PuGPu)] alkylation in the minor groove of DNA through covalent binding from guanine N2 to the PBD C11-position. Dimerization of the PBD ring system gives molecules that can cross-link DNA, which leads to a substantial increase in potency and DNA binding affinity and an extension of sequence-selectivity compared with monomers. PBD dimers feature as the cytotoxic component of numerous ADCs being evaluated in clinical trials. PBD-ADC clinical candidates loncastuximab tesirine, camidanlumab tesirine and rovalpituzumab tesirine employ a PBD N10 linkage while vadastuximab talirine uses a C2-linkage. The PBD dimer scaffold is versatile and offers many opportunities to diversify the ADC platform, with extensive research being performed worldwide to develop the next generation of PBD payload–linker molecules. The search for new PBD payload–linker molecules has mainly focused on changes in payload structure (e.g. PBD C2 modification and macrocyclisation), alternative conjugation strategies (e.g. haloacetamides, ‘click’ approaches and pyridyl disulphides), non-peptide triggers in the linker (e.g. disulphides) and non-cleavable derivatives (i.e. payload release through antibody degradation).
Collapse
|
21
|
MacGregor P, Gonzalez-Munoz AL, Jobe F, Taylor MC, Rust S, Sandercock AM, Macleod OJS, Van Bocxlaer K, Francisco AF, D’Hooge F, Tiberghien A, Barry CS, Howard P, Higgins MK, Vaughan TJ, Minter R, Carrington M. A single dose of antibody-drug conjugate cures a stage 1 model of African trypanosomiasis. PLoS Negl Trop Dis 2019; 13:e0007373. [PMID: 31120889 PMCID: PMC6532856 DOI: 10.1371/journal.pntd.0007373] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/09/2019] [Indexed: 02/02/2023] Open
Abstract
Infections of humans and livestock with African trypanosomes are treated with drugs introduced decades ago that are not always fully effective and often have severe side effects. Here, the trypanosome haptoglobin-haemoglobin receptor (HpHbR) has been exploited as a route of uptake for an antibody-drug conjugate (ADC) that is completely effective against Trypanosoma brucei in the standard mouse model of infection. Recombinant human anti-HpHbR monoclonal antibodies were isolated and shown to be internalised in a receptor-dependent manner. Antibodies were conjugated to a pyrrolobenzodiazepine (PBD) toxin and killed T. brucei in vitro at picomolar concentrations. A single therapeutic dose (0.25 mg/kg) of a HpHbR antibody-PBD conjugate completely cured a T. brucei mouse infection within 2 days with no re-emergence of infection over a subsequent time course of 77 days. These experiments provide a demonstration of how ADCs can be exploited to treat protozoal diseases that desperately require new therapeutics.
Collapse
Affiliation(s)
- Paula MacGregor
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | | | - Fatoumatta Jobe
- Department of Antibody Discovery and Protein Engineering, Medimmune, Cambridge, United Kingdom
| | - Martin C. Taylor
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Steven Rust
- Department of Antibody Discovery and Protein Engineering, Medimmune, Cambridge, United Kingdom
| | - Alan M. Sandercock
- Department of Antibody Discovery and Protein Engineering, Medimmune, Cambridge, United Kingdom
| | | | | | | | | | | | | | | | - Matthew K. Higgins
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Tristan J. Vaughan
- Department of Antibody Discovery and Protein Engineering, Medimmune, Cambridge, United Kingdom
| | - Ralph Minter
- Department of Antibody Discovery and Protein Engineering, Medimmune, Cambridge, United Kingdom
| | - Mark Carrington
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
22
|
Li Q, Barrett A, Vijayakrishnan B, Tiberghien A, Beard R, Rickert KW, Allen KL, Christie RJ, Marelli M, Harper J, Howard P, Wu H, Dall'Acqua WF, Tsui P, Gao C, Borrok MJ. Improved Inhibition of Tumor Growth by Diabody-Drug Conjugates via Half-Life Extension. Bioconjug Chem 2019; 30:1232-1243. [PMID: 30912649 DOI: 10.1021/acs.bioconjchem.9b00170] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Despite some clinical success with antibody-drug conjugates (ADCs) in patients with solid tumors and hematological malignancies, improvements in ADC design are still desirable due to the narrow therapeutic window of these compounds. Tumor-targeting antibody fragments have distinct advantages over monoclonal antibodies, including more rapid tumor accumulation and enhanced penetration, but are subject to rapid clearance. Half-life extension technologies such as PEGylation and albumin-binding domains (ABDs) have been widely used to improve the pharmacokinetics of many different types of biologics. PEGylation improves pharmacokinetics by increasing hydrodynamic size to reduce renal clearance, whereas ABDs extend half-life via FcRn-mediated recycling. In this study, we used an anti-oncofetal antigen 5T4 diabody conjugated with a highly potent cytotoxic pyrrolobenzodiazepine (PBD) warhead to assess and compare the effects of PEGylation and albumin binding on the in vivo efficacy of antibody fragment drug conjugates. Conjugation of 2× PEG20K to a diabody improved half-life from 40 min to 33 h, and an ABD-diabody fusion protein exhibited a half-life of 45 h in mice. In a xenograft model of breast cancer MDA-MB-436, the ABD-diabody-PBD showed greater tumor growth suppression and better tolerability than either PEG-diabody-PBD or diabody-PBD. These results suggest that the mechanism of half-life extension is an important consideration for designing cytotoxic antitumor agents.
Collapse
Affiliation(s)
| | | | | | | | - Rhiannon Beard
- Spirogen , 42 New Road , E1 2AX , London , United Kingdom
| | | | | | | | | | | | - Philip Howard
- Spirogen , 42 New Road , E1 2AX , London , United Kingdom
| | | | | | | | | | | |
Collapse
|
23
|
Shi B, Wu M, Li Z, Xie Z, Wei X, Fan J, Xu Y, Ding D, Akash SH, Chen S, Cao S. Antitumor activity of a 5T4 targeting antibody drug conjugate with a novel payload derived from MMAF via C-Lock linker. Cancer Med 2019; 8:1793-1805. [PMID: 30843650 PMCID: PMC6488119 DOI: 10.1002/cam4.2066] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/15/2019] [Accepted: 02/12/2019] [Indexed: 12/19/2022] Open
Abstract
Antibody-drug conjugates (ADCs) belong to a promising class of biopharmaceuticals in which target-killing of tumor cells was achieved by marrying the potency of the cytotoxic payload with the tumor specificity of the antibody. Here we developed a novel ADC (ZV0508) that targets 5T4 oncofetal antigen, which is overexpressed in many carcinomas on both bulk tumor cells and cancer stem cells. A novel cytotoxic payload called Duostatin-5 (Duo-5) which was derived from monomethyl auristatin F (MMAF) was attached to a 5T4 targeting antibody (ZV05) by interchain cysteine cross-linking conjugation via a disubstituted C-Lock linker. We have investigated the antitumor efficacy of ZV0508 by in vitro and in vivo studies, and compared its antitumor activity with ZV05-mcMMAF (ZV0501), in which MMAF was linked via a conventional noncleavable maleimidocaproyl linker. As results, ZV0508 exhibited ideal antiproliferative effects through blocking cell cycle and inducing cell apoptosis. The in vivo studies revealed that both ZV0501 and ZV0508 exhibited excellent antitumor activities even at a single dose. Although ZV0508 was inferior to ZV0501 in vitro, it elicited more durable antitumor responses than ZV0501 in vivo. The superior in vivo activity of ZV0508 may be due to the combined use of the disubstituted C-Lock linker and the novel payload Duo-5, resulting in a more stable and potent ADC. Taken together, these data suggest ZV0508 is a worthy candidate for the treatment of 5T4 positive cancers.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antigens, Neoplasm/metabolism
- Antineoplastic Agents/pharmacokinetics
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Apoptosis/drug effects
- Cell Cycle Checkpoints/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cross-Linking Reagents
- Female
- Humans
- Immunoconjugates/pharmacokinetics
- Immunoconjugates/pharmacology
- Immunoconjugates/therapeutic use
- Male
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mice, Inbred BALB C
- Mice, Nude
- Molecular Targeted Therapy/methods
- Neoplasms/drug therapy
- Neoplasms/metabolism
- Neoplasms/pathology
- Oligopeptides/pharmacology
- Tumor Cells, Cultured/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Baoying Shi
- College of Pharmaceutical Sciences, Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Provincial Key Laboratory of Anti‐Cancer Drug ResearchZhejiang UniversityHangzhouChina
| | - Min Wu
- Zova Biotherapeutics IncFuyang, HangzhouChina
| | - Zhaohui Li
- Zova Biotherapeutics IncFuyang, HangzhouChina
| | | | - Xiaoyue Wei
- College of Pharmaceutical Sciences, Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Provincial Key Laboratory of Anti‐Cancer Drug ResearchZhejiang UniversityHangzhouChina
| | - Jiansheng Fan
- College of Pharmaceutical Sciences, Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Provincial Key Laboratory of Anti‐Cancer Drug ResearchZhejiang UniversityHangzhouChina
| | - Yingchun Xu
- College of Pharmaceutical Sciences, Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Provincial Key Laboratory of Anti‐Cancer Drug ResearchZhejiang UniversityHangzhouChina
| | - Ding Ding
- Noeantigen Therapeutics (HangZhou) Co., LtdHangzhouChina
| | - Sajid Hamid Akash
- College of Pharmaceutical Sciences, Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Provincial Key Laboratory of Anti‐Cancer Drug ResearchZhejiang UniversityHangzhouChina
| | - Shuqing Chen
- College of Pharmaceutical Sciences, Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Provincial Key Laboratory of Anti‐Cancer Drug ResearchZhejiang UniversityHangzhouChina
| | - Sheldon Cao
- Zova Biotherapeutics IncFuyang, HangzhouChina
| |
Collapse
|
24
|
Marcucci F, Caserta CA, Romeo E, Rumio C. Antibody-Drug Conjugates (ADC) Against Cancer Stem-Like Cells (CSC)-Is There Still Room for Optimism? Front Oncol 2019; 9:167. [PMID: 30984612 PMCID: PMC6449442 DOI: 10.3389/fonc.2019.00167] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 02/25/2019] [Indexed: 01/06/2023] Open
Abstract
Cancer stem-like cells (CSC) represent a subpopulation of tumor cells with peculiar functionalities that distinguish them from the bulk of tumor cells, most notably their tumor-initiating potential and drug resistance. Given these properties, it appears logical that CSCs have become an important target for many pharma companies. Antibody-drug conjugates (ADC) have emerged over the last decade as one of the most promising new tools for the selective ablation of tumor cells. Three ADCs have already received regulatory approval and many others are in different phases of clinical development. Not surprisingly, also a considerable number of anti-CSC ADCs have been described in the literature and some of these have entered clinical development. Several of these ADCs, however, have yielded disappointing results in clinical studies. This is similar to the results obtained with other anti-CSC drug candidates, including native antibodies, that have been investigated in the clinic. In this article we review the anti-CSC ADCs that have been described in the literature and, in the following, we discuss reasons that may underlie the failures in clinical trials that have been observed. Possible reasons relate to the biology of CSCs themselves, including their heterogeneity, the lack of strictly CSC-specific markers, and the capacity to interconvert between CSCs and non-CSCs; second, inherent limitations of some classes of cytotoxins that have been used for the construction of ADCs; third, the inadequacy of animal models in predicting efficacy in humans. We conclude suggesting some possibilities to address these limitations.
Collapse
Affiliation(s)
- Fabrizio Marcucci
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | | | | | - Cristiano Rumio
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| |
Collapse
|
25
|
White JB, Fleming R, Masterson L, Ruddle BT, Zhong H, Fazenbaker C, Strout P, Rosenthal K, Reed M, Muniz-Medina V, Howard P, Dixit R, Wu H, Hinrichs MJ, Gao C, Dimasi N. Design and characterization of homogenous antibody-drug conjugates with a drug-to-antibody ratio of one prepared using an engineered antibody and a dual-maleimide pyrrolobenzodiazepine dimer. MAbs 2019; 11:500-515. [PMID: 30835621 DOI: 10.1080/19420862.2019.1578611] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Most strategies used to prepare homogeneous site-specific antibody-drug conjugates (ADCs) result in ADCs with a drug-to-antibody ratio (DAR) of two. Here, we report a disulfide re-bridging strategy to prepare homogeneous ADCs with DAR of one using a dual-maleimide pyrrolobenzodiazepine (PBD) dimer (SG3710) and an engineered antibody (Flexmab), which has only one intrachain disulfide bridge at the hinge. We demonstrate that SG3710 efficiently re-bridge a Flexmab targeting human epidermal growth factor receptor 2 (HER2), and the resulting ADC was highly resistant to payload loss in serum and exhibited potent anti-tumor activity in a HER2-positive gastric carcinoma xenograft model. Moreover, this ADC was tolerated in rats at twice the dose compared to a site-specific ADC with DAR of two prepared using a single-maleimide PBD dimer (SG3249). Flexmab technologies, in combination with SG3710, provide a platform for generating site-specific homogenous PBD-based ADCs with DAR of one, which have improved biophysical properties and tolerability compared to conventional site-specific PBD-based ADCs with DAR of two.
Collapse
Affiliation(s)
- Jason B White
- a Antibody Discovery and Protein Engineering , MedImmune , Gaithersburg , MD , USA
| | - Ryan Fleming
- a Antibody Discovery and Protein Engineering , MedImmune , Gaithersburg , MD , USA
| | | | - Ben T Ruddle
- a Antibody Discovery and Protein Engineering , MedImmune , Gaithersburg , MD , USA
| | - Haihong Zhong
- c Oncology Research , MedImmune , Gaithersburg , MD , USA
| | | | - Patrick Strout
- c Oncology Research , MedImmune , Gaithersburg , MD , USA
| | - Kim Rosenthal
- a Antibody Discovery and Protein Engineering , MedImmune , Gaithersburg , MD , USA
| | - Molly Reed
- d Biologics Safety Assessment , MedImmune , Gaithersburg , MD , USA
| | | | - Philip Howard
- b Spirogen Ltd , QMB Innovation Center , London , UK
| | - Rakesh Dixit
- d Biologics Safety Assessment , MedImmune , Gaithersburg , MD , USA
| | - Herren Wu
- a Antibody Discovery and Protein Engineering , MedImmune , Gaithersburg , MD , USA
| | | | - Changshou Gao
- a Antibody Discovery and Protein Engineering , MedImmune , Gaithersburg , MD , USA
| | - Nazzareno Dimasi
- a Antibody Discovery and Protein Engineering , MedImmune , Gaithersburg , MD , USA
| |
Collapse
|
26
|
Harrop R, O’Neill E, Stern PL. Cancer stem cell mobilization and therapeutic targeting of the 5T4 oncofetal antigen. Ther Adv Vaccines Immunother 2019; 7:2515135518821623. [PMID: 30719508 PMCID: PMC6348545 DOI: 10.1177/2515135518821623] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 11/27/2018] [Indexed: 12/21/2022] Open
Abstract
Cancer stem cells (CSCs) can act as the cellular drivers of tumors harnessing stem cell properties that contribute to tumorigenesis either as founder elements or by the gain of stem cell traits by the malignant cells. Thus, CSCs can self-renew and generate the cellular heterogeneity of tumors including a hierarchical organization similar to the normal tissue. While the principle tumor growth contribution is often from the non-CSC components, it is the ability of small numbers of CSCs to avoid the effects of therapeutic strategies that can contribute to recurrence after treatment. However, identifying and characterizing CSCs for therapeutic targeting is made more challenging by their cellular potency being influenced by a particular tissue niche or by the capacity of more committed cells to regain stem cell functions. This review discusses the properties of CSCs including the limitations of the available cell surface markers, the assays that document tumor initiation and clonogenicity, the roles of epithelial mesenchymal transition and molecular pathways such as Notch, Wnt, Hippo and Hedgehog. The ability to target and eliminate CSCs is thought to be critical in the search for curative cancer treatments. The oncofetal tumor-associated antigen 5T4 (TBGP) has been linked with CSC properties in several different malignancies. 5T4 has functional attributes that are relevant to the spread of tumors including through EMT, CXCR4/CXCL12, Wnt, and Hippo pathways which may all contribute through the mobilization of CSCs. There are several different immunotherapies targeting 5T4 in development including antibody-drug conjugates, antibody-targeted bacterial super-antigens, a Modified Vaccinia Ankara-basedvaccine and 5T4-directed chimeric antigen receptor T-cells. These immune therapies would have the advantage of targeting both the bulk tumor as well as mobilized CSC populations.
Collapse
Affiliation(s)
- Richard Harrop
- Oxford BioMedica plc, Windrush Court, Transport Way, Oxford, OX4 6LT, UK
| | - Eric O’Neill
- Department of Oncology, University of Oxford, Oxford, UK
| | - Peter L. Stern
- Division of Molecular & Clinical Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| |
Collapse
|
27
|
Khera E, Thurber GM. Pharmacokinetic and Immunological Considerations for Expanding the Therapeutic Window of Next-Generation Antibody-Drug Conjugates. BioDrugs 2019; 32:465-480. [PMID: 30132210 DOI: 10.1007/s40259-018-0302-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Antibody-drug conjugate (ADC) development has evolved greatly over the last 3 decades, including the Food and Drug Administration (FDA) approval of several new drugs. However, translating ADCs from the design stage and preclinical promise to clinical success has been a major hurdle for the field, particularly for solid tumors. The challenge in clinical development can be attributed to the difficulty in connecting the design of these multifaceted agents with the impact on clinical efficacy, especially with the accelerated development of 'next-generation' ADCs containing a variety of innovative biophysical developments. Given their complex nature, there is an urgent need to integrate holistic ADC characterization approaches. This includes comprehensive in vivo assessment of systemic, intratumoral and cellular pharmacokinetics, pharmacodynamics, toxicodynamics, and interactions with the immune system, with the aim of optimizing the ADC therapeutic window. Pharmacokinetic/pharmacodynamic factors influencing the ADC therapeutic window include (1) selecting optimal target and ADC components for prolonged and stable plasma circulation to increase tumoral uptake with minimal non-specific systemic toxicity, (2) balancing homogeneous intratumoral distribution with efficient cellular uptake, and (3) translating improved ADC potency to better clinical efficacy. Balancing beneficial immunological effects such as Fc-mediated and payload-mediated immune cell activation against harmful immunogenic/toxic effects is also an emerging concern for ADCs. Here, we review practical considerations for tracking ADC efficacy and toxicity, as aided by high-resolution biomolecular and immunological tools, quantitative pharmacology, and mathematical models, all of which can elucidate the relative contributions of the multitude of interactions governing the ADC therapeutic window.
Collapse
Affiliation(s)
- Eshita Khera
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Rd, Ann Arbor, MI, 48109, USA
| | - Greg M Thurber
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Rd, Ann Arbor, MI, 48109, USA. .,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
28
|
Farouk F, Shamma R. Chemical structure modifications and nano-technology applications for improving ADME-Tox properties, a review. Arch Pharm (Weinheim) 2019; 352:e1800213. [DOI: 10.1002/ardp.201800213] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 11/02/2018] [Accepted: 11/11/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Faten Farouk
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry; Ahram Canadian University; Giza Egypt
| | - Rehab Shamma
- Faculty of Pharmacy, Department of Pharmaceutics and Industrial Pharmacy; Cairo University; Cairo Egypt
| |
Collapse
|
29
|
ADME Considerations and Bioanalytical Strategies for Pharmacokinetic Assessments of Antibody-Drug Conjugates. Antibodies (Basel) 2018; 7:antib7040041. [PMID: 31544891 PMCID: PMC6698957 DOI: 10.3390/antib7040041] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/21/2018] [Accepted: 11/26/2018] [Indexed: 12/19/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are a unique class of biotherapeutics of inherent heterogeneity and correspondingly complex absorption, distribution, metabolism, and excretion (ADME) properties. Herein, we consider the contribution of various components of ADCs such as various classes of warheads, linkers, and conjugation strategies on ADME of ADCs. Understanding the metabolism and disposition of ADCs and interpreting exposure-efficacy and exposure-safety relationships of ADCs in the context of their various catabolites is critical for design and subsequent development of a clinically successful ADCs. Sophisticated bioanalytical assays are required for the assessments of intact ADC, total antibody, released warhead and relevant metabolites. Both ligand-binding assays (LBA) and hybrid LBA-liquid chromatography coupled with tandem mass spectrometry (LBA-LC-MS/MS) methods have been employed to assess pharmacokinetics (PK) of ADCs. Future advances in bioanalytical techniques will need to address the rising complexity of this biotherapeutic modality as more innovative conjugation strategies, antibody scaffolds and novel classes of warheads are employed for the next generation of ADCs. This review reflects our considerations on ADME of ADCs and provides a perspective on the current bioanalytical strategies for pharmacokinetic assessments of ADCs.
Collapse
|
30
|
Zhong H, Chen C, Tammali R, Breen S, Zhang J, Fazenbaker C, Kennedy M, Conway J, Higgs BW, Holoweckyj N, Raja R, Harper J, Pierce AJ, Herbst R, Tice DA. Improved Therapeutic Window in BRCA-mutant Tumors with Antibody-linked Pyrrolobenzodiazepine Dimers with and without PARP Inhibition. Mol Cancer Ther 2018; 18:89-99. [DOI: 10.1158/1535-7163.mct-18-0314] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 07/31/2018] [Accepted: 10/16/2018] [Indexed: 11/16/2022]
|
31
|
Preclinical assessment of an antibody–PBD conjugate that targets BCMA on multiple myeloma and myeloma progenitor cells. Leukemia 2018; 33:766-771. [DOI: 10.1038/s41375-018-0278-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/27/2018] [Accepted: 09/03/2018] [Indexed: 01/10/2023]
|
32
|
Tiberghien AC, von Bulow C, Barry C, Ge H, Noti C, Collet Leiris F, McCormick M, Howard PW, Parker JS. Scale-up Synthesis of Tesirine. Org Process Res Dev 2018. [DOI: 10.1021/acs.oprd.8b00205] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
| | | | - Conor Barry
- Spirogen, QMB Innovation Centre, 42 New Road, E1 2AX London, United Kingdom
| | - Huajun Ge
- Pharmaron, No. 6, Taihe Road, BDA, Beijing, 100176, People’s Republic of China
| | | | | | - Marc McCormick
- Early Chemical Development, Pharmaceutical Sciences, IMED Biotech Unit, AstraZeneca, Macclesfield, United Kingdom
| | - Philip W. Howard
- Spirogen, QMB Innovation Centre, 42 New Road, E1 2AX London, United Kingdom
| | - Jeremy S. Parker
- Early Chemical Development, Pharmaceutical Sciences, IMED Biotech Unit, AstraZeneca, Macclesfield, United Kingdom
| |
Collapse
|
33
|
Glucuronide-Linked Antibody–Tubulysin Conjugates Display Activity in MDR+ and Heterogeneous Tumor Models. Mol Cancer Ther 2018; 17:1752-1760. [DOI: 10.1158/1535-7163.mct-18-0073] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/06/2018] [Accepted: 05/18/2018] [Indexed: 11/16/2022]
|
34
|
Tumor uptake of pegylated diabodies: Balancing systemic clearance and vascular transport. J Control Release 2018; 279:126-135. [DOI: 10.1016/j.jconrel.2018.04.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 03/29/2018] [Accepted: 04/06/2018] [Indexed: 01/06/2023]
|
35
|
Bosco EE, Christie RJ, Carrasco R, Sabol D, Zha J, DaCosta K, Brown L, Kennedy M, Meekin J, Phipps S, Ayriss J, Du Q, Bezabeh B, Chowdhury P, Breen S, Chen C, Reed M, Hinrichs M, Zhong H, Xiao Z, Dixit R, Herbst R, Tice DA. Preclinical evaluation of a GFRA1 targeted antibody-drug conjugate in breast cancer. Oncotarget 2018; 9:22960-22975. [PMID: 29796165 PMCID: PMC5955426 DOI: 10.18632/oncotarget.25160] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/05/2018] [Indexed: 02/07/2023] Open
Abstract
Despite recent advances in treatment, breast cancer remains the second-most common cause of cancer death among American women. A greater understanding of the molecular characteristics of breast tumors could ultimately lead to improved tumor-targeted treatment options, particularly for subsets of breast cancer patients with unmet needs. Using an unbiased genomics approach to uncover membrane-localized tumor-associated antigens (TAAs), we have identified glial cell line derived neurotrophic factor (GDNF) family receptor α 1 (GFRA1) as a breast cancer TAA. Immunohistochemistry (IHC) revealed that GFRA1 displays a limited normal tissue expression profile coupled with overexpression in specific breast cancer subsets. The cell surface localization as determined by fluorescence-activated cell sorting (FACS) and the rapid internalization kinetics of GFRA1 makes it an ideal target for therapeutic exploitation as an antibody-drug conjugate (ADC). Here, we describe the development of a pyrrolobenzodiazepine (PBD)-armed, GFRA1-targeted ADC that demonstrates cytotoxicity in GFRA1-positive cell lines and patient-derived xenograft (PDX) models. The safety profile of the rat cross-reactive GFRA1-PBD was assessed in a rat toxicology study to find transient cellularity reductions in the bone marrow and peripheral blood, consistent with known off-target effects of PBD ADC's. These studies reveal no evidence of on-target toxicity and support further evaluation of GFRA1-PBD in GFRA1-positive tumors.
Collapse
Affiliation(s)
- Emily E. Bosco
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland, United States of America
| | - R. James Christie
- Antibody Discovery and Protein Engineering, MedImmune, LLC, Gaithersburg, Maryland, United States of America
| | - Rosa Carrasco
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland, United States of America
| | - Darrin Sabol
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland, United States of America
| | - Jiping Zha
- Pathology, MedImmune, LLC, Gaithersburg, Maryland, United States of America
- Translational Sciences, NGM Biopharmaceuticals, South San Francisco, California, United States of America
| | - Karma DaCosta
- Pathology, MedImmune, LLC, Gaithersburg, Maryland, United States of America
| | - Lee Brown
- Pathology, MedImmune, Ltd, Cambridge, United Kingdom
| | - Maureen Kennedy
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland, United States of America
| | - John Meekin
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland, United States of America
| | - Sandrina Phipps
- Antibody Discovery and Protein Engineering, MedImmune, LLC, Gaithersburg, Maryland, United States of America
| | - Joanne Ayriss
- Antibody Discovery and Protein Engineering, MedImmune, LLC, Gaithersburg, Maryland, United States of America
- Department of Global Biotherapeutics, Pfizer, Cambridge, Massachusetts, United States of America
| | - Qun Du
- Antibody Discovery and Protein Engineering, MedImmune, LLC, Gaithersburg, Maryland, United States of America
| | - Binyam Bezabeh
- Antibody Discovery and Protein Engineering, MedImmune, LLC, Gaithersburg, Maryland, United States of America
- Research, Salubris Biotherapeutics, Gaithersburg, Maryland, United States of America
| | - Partha Chowdhury
- Antibody Discovery and Protein Engineering, MedImmune, LLC, Gaithersburg, Maryland, United States of America
- Biologics Discovery, Sanofi Genzyme, Cambridge, MA, United States of America
| | - Shannon Breen
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland, United States of America
| | - Cui Chen
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland, United States of America
| | - Molly Reed
- Biologics Safety Assessment, MedImmune, LLC, Gaithersburg, Maryland, United States of America
| | - MaryJane Hinrichs
- Biologics Safety Assessment, MedImmune, LLC, Gaithersburg, Maryland, United States of America
| | - Haihong Zhong
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland, United States of America
| | - Zhan Xiao
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland, United States of America
| | - Rakesh Dixit
- Biologics Safety Assessment, MedImmune, LLC, Gaithersburg, Maryland, United States of America
| | - Ronald Herbst
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland, United States of America
| | - David A. Tice
- Oncology Research, MedImmune, LLC, Gaithersburg, Maryland, United States of America
| |
Collapse
|
36
|
Miller ML, Shizuka M, Wilhelm A, Salomon P, Reid EE, Lanieri L, Sikka S, Maloney EK, Harvey L, Qiu Q, Archer KE, Bai C, Vitharana D, Harris L, Singh R, Ponte JF, Yoder NC, Kovtun Y, Lai KC, Ab O, Pinkas J, Keating TA, Chari RV. A DNA-Interacting Payload Designed to Eliminate Cross-Linking Improves the Therapeutic Index of Antibody–Drug Conjugates (ADCs). Mol Cancer Ther 2018; 17:650-660. [DOI: 10.1158/1535-7163.mct-17-0940] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 11/15/2017] [Accepted: 12/28/2017] [Indexed: 11/16/2022]
|
37
|
Christie RJ, Tiberghien AC, Du Q, Bezabeh B, Fleming R, Shannon A, Mao S, Breen S, Zhang J, Zhong H, Harper J, Wu H, Howard PW, Gao C. Pyrrolobenzodiazepine Antibody-Drug Conjugates Designed for Stable Thiol Conjugation. Antibodies (Basel) 2017; 6:antib6040020. [PMID: 31548535 PMCID: PMC6698857 DOI: 10.3390/antib6040020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/20/2017] [Accepted: 11/21/2017] [Indexed: 12/24/2022] Open
Abstract
Thiosuccinimide-linked antibody-drug conjugates (ADCs) are susceptible to drug loss over time due to a retro-Michael reaction, which can be prevented by selecting stable conjugation positions or hydrolysis of the thiosuccinimide. Here, we investigate pyrrolobenzodiazepine (PBD) ADC drug-linkers equipped with N-phenyl maleimide functionality for stable thiol conjugation via thiosuccinimide hydrolysis. Two PBD drug-linker formats (enzyme-cleavable and non-cleavable) were evaluated following site-specific conjugation to an engineered cysteine incorporated at position T289, which is known to be unstable for N-alkyl maleimide conjugates. N-phenyl maleimide PBDs conjugated to antibodies with similar efficiencies as N-alkyl maleimide PBDs and enhanced thiosuccinimide hydrolysis for N-phenyl maleimide PBDs was confirmed by mass spectrometry, capillary isoelectric focusing, and a SYPRO Orange dye binding assay. All of the PBD ADCs were highly potent in vitro regardless of maleimide- or linker-type, exhibiting low pM EC50 values. Thiol conjugation to N-phenyl maleimide PBD minimized the retro-Michael reaction in both rat and mouse serum. However, cleavage of the valine-alanine dipeptide in mouse serum for ADCs containing cleavable drug-linker led to drug loss regardless of maleimide type, which impacted ADC potency in tumor growth inhibition studies that were conducted in mouse models. Therapeutic improvement in mouse tumor models was realized for ADCs prepared with non-cleavable PBD drug-linkers that were conjugated through N-phenyl maleimide, where a stronger tumor growth inhibition (TGI) response was achieved when compared to the analogous N-alkyl maleimide drug-linker ADC. Altogether, our findings highlight the stability and efficacy benefits of N-phenyl maleimide functionality for ADCs that are produced with thiol-maleimide conjugation chemistry.
Collapse
Affiliation(s)
- R James Christie
- Antibody Discovery and Protein Engineering, MedImmune, Gaithersburg, MD 20878, USA.
| | | | - Qun Du
- Antibody Discovery and Protein Engineering, MedImmune, Gaithersburg, MD 20878, USA.
| | - Binyam Bezabeh
- Antibody Discovery and Protein Engineering, MedImmune, Gaithersburg, MD 20878, USA.
| | - Ryan Fleming
- Antibody Discovery and Protein Engineering, MedImmune, Gaithersburg, MD 20878, USA.
| | - Amanda Shannon
- Antibody Discovery and Protein Engineering, MedImmune, Gaithersburg, MD 20878, USA.
| | - Shenlan Mao
- Oncology Research, MedImmune, Gaithersburg, MD 20878, USA.
| | - Shannon Breen
- Oncology Research, MedImmune, Gaithersburg, MD 20878, USA.
| | - Jing Zhang
- Oncology Research, MedImmune, Gaithersburg, MD 20878, USA.
| | - Haihong Zhong
- Oncology Research, MedImmune, Gaithersburg, MD 20878, USA.
| | - Jay Harper
- Oncology Research, MedImmune, Gaithersburg, MD 20878, USA.
| | - Herren Wu
- Antibody Discovery and Protein Engineering, MedImmune, Gaithersburg, MD 20878, USA.
| | | | - Changshou Gao
- Antibody Discovery and Protein Engineering, MedImmune, Gaithersburg, MD 20878, USA.
| |
Collapse
|