1
|
Moore EJ, Rice M, Roy G, Zhang W, Marelli M. Emerging conjugation strategies and protein engineering technologies aim to improve ADCs in the fight against cancer. Xenobiotica 2024; 54:469-491. [PMID: 39329289 DOI: 10.1080/00498254.2024.2339993] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/22/2024] [Accepted: 04/03/2024] [Indexed: 09/28/2024]
Abstract
Antibody drug conjugates are an exciting therapeutic modality that combines the targeting specificity of antibodies with potent cytotoxins to selectively kill cancer cells. The targeting component improves efficacy and protects non-target cells from the harmful effects of the payload. To date 15 ADCs have been approved by regulatory agencies for commercial use and shown to be valuable tools in the treatment of cancer.The assembly of an ADC requires the chemical ligation of a linker-payload to an antibody. Conventional conjugation methods targeting accessible lysines and cysteines have produced all the ADCs currently on the market. While successful, technologies aiming to improve the homogeneity and stability of ADCs are being developed and tested.Here we provide a review of developing methods for ADC construction. These include enzymatic methods, oligosaccharide remodelling, and technologies using genetic code expansion techniques. The virtues and limitations of each technology are discussed.Emerging conjugation technologies are being applied to produce new formats of ADCs with enhanced functionality including bispecific ADCs, dual-payload ADCs, and nanoparticles for targeted drug delivery. The benefits of these novel formats are highlighted.
Collapse
|
2
|
Lindgren ES, Yan R, Cil O, Verkman AS, Chan MF, Seitzman GD, Farooq AV, Huppert LA, Rugo HS, Pohlmann PR, Lu J, Esserman LJ, Pasricha ND. Incidence and Mitigation of Corneal Pseudomicrocysts Induced by Antibody-Drug Conjugates (ADCs). CURRENT OPHTHALMOLOGY REPORTS 2024; 12:13-22. [PMID: 38756824 PMCID: PMC11095972 DOI: 10.1007/s40135-024-00322-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2024] [Indexed: 05/18/2024]
Abstract
Purpose of Review This study is to highlight the incidence of corneal pseudomicrocysts in FDA-approved antibody-drug conjugates (ADCs), and success of preventive therapies for pseudomicrocysts and related ocular surface adverse events (AEs). Recent Findings ADCs are an emerging class of selective cancer therapies that consist of a potent cytotoxin connected to a monoclonal antibody (mAb) that targets antigens expressed on malignant cells. Currently, there are 11 FDA-approved ADCs with over 164 in clinical trials. Various AEs have been attributed to ADCs, including ocular surface AEs (keratitis/keratopathy, dry eye, conjunctivitis, blurred vision, corneal pseudomicrocysts). While the severity and prevalence of ADC-induced ocular surface AEs are well reported, the reporting of corneal pseudomicrocysts is limited, complicating the development of therapies to prevent or treat ADC-related ocular surface toxicity. Summary Three of 11 FDA-approved ADCs have been implicated with corneal pseudomicrocysts, with incidence ranging from 41 to 100% of patients. Of the six ADCs that reported ocular surface AEs, only three had ocular substudies to investigate the benefit of preventive therapies including topical steroids, vasoconstrictors, and preservative-free lubricants. Current preventive therapies demonstrate limited efficacy at mitigating pseudomicrocysts and other ocular surface AEs.
Collapse
Affiliation(s)
- Ethan S. Lindgren
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, USA
| | - Rongshan Yan
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, USA
| | - Onur Cil
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Alan S. Verkman
- Departments of Medicine and Physiology, University of California San Francisco, San Francisco, CA, USA
| | - Matilda F. Chan
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, USA
- Francis I. Proctor Foundation, University of California San Francisco, San Francisco, CA, USA
| | - Gerami D. Seitzman
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, USA
- Francis I. Proctor Foundation, University of California San Francisco, San Francisco, CA, USA
| | - Asim V. Farooq
- Department of Ophthalmology and Visual Science, University of Chicago Medical Center, Chicago, IL, USA
| | - Laura A. Huppert
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Hope S. Rugo
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Paula R. Pohlmann
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Janice Lu
- Department of Medical Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Laura J. Esserman
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Neel D. Pasricha
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, USA
- Francis I. Proctor Foundation, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
3
|
Marcelletti JF, Sikic BI. A clinical trial of zosuquidar plus gemtuzumab ozogamicin (GO) in relapsed or refractory acute myeloid leukemia (RR AML): evidence of efficacy based on leukemic blast P-glycoprotein functional phenotype. Cancer Chemother Pharmacol 2023; 92:369-380. [PMID: 37603048 DOI: 10.1007/s00280-023-04578-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/05/2023] [Indexed: 08/22/2023]
Abstract
PURPOSE To evaluate safety, tolerability, potential efficacy, and pharmacodynamics (PD) of zosuquidar (Zos) in combination with gemtuzumab ozogamicin (GO) in elderly patients with relapsed or refractory (RR) acute myeloid leukemia (AML). METHODS Patients with RR AML (N = 41) were treated with Zos as a 48-h continuous intravenous infusion initiated 4 h prior to a 2-h infusion of GO on days 1 and 15. P-glycoprotein (P-gp) status of the patients' leukemic blasts and PD determinations were assessed with ex vivo bioassays. Patient outcomes were analyzed for the total cohort and as stratified into P-gp-positive (P-gp +) and P-gp-negative (P-gp‒) subgroups. RESULTS The eligible cohort exhibited a 34% overall remission rate (ORR), a composite of patients that exhibited complete remission (CR), CR with incomplete platelet recovery, or morphologic remission. Patients with 1st relapsed disease exhibited 40% ORR. P-gp phenotype did not significantly predict ORR. However, the P-gp + subgroup exhibited a greater median overall survival (OS) of 6.0 months vs. 1.8 months for patients in the P-gp‒ subgroup (p = 0.01). PD analyses revealed 90-95% inhibition of blast P-gp function during Zos infusion. Treatment related toxicities were observed and resolved with decrease or discontinued Zos or GO dosages. CONCLUSIONS Zos plus GO elicited appreciable ORR for an elderly patient population with RR AML. The greater OS of the P-gp + subgroup vs. the P-gp‒ subgroup suggests that patients with P-gp + leukemic blasts were being more effectively targeted by GO with Zos co-therapy. The poorer OS of the P-gp‒ subgroup suggests activity of Zos-insensitive multidrug resistant mechanisms. CLINICAL TRIALS GOV IDENTIFIER NCT00233909; First posted October 06, 2005.
Collapse
Affiliation(s)
| | - Branimir I Sikic
- Kanisa Pharmaceuticals, San Diego, CA, USA
- Clinical and Translational Research Unit, Stanford Cancer Institute, Stanford, CA, USA
| |
Collapse
|
4
|
Antibody-Drug Conjugates in Myeloid Leukemias. Cancer J 2022; 28:454-461. [DOI: 10.1097/ppo.0000000000000635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
5
|
High-Risk Acute Myeloid Leukemia: A Pediatric Prospective. Biomedicines 2022; 10:biomedicines10061405. [PMID: 35740427 PMCID: PMC9220202 DOI: 10.3390/biomedicines10061405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/19/2022] [Accepted: 05/31/2022] [Indexed: 11/16/2022] Open
Abstract
Pediatric acute myeloid leukemia is a clonal disorder characterized by malignant transformation of the hematopoietic stem cell. The incidence and the outcome remain inferior when compared to pediatric ALL, although prognosis has improved in the last decades, with 80% overall survival rate reported in some studies. The standard therapeutic approach is a combined cytarabine and anthracycline-based regimen followed by consolidation with allogeneic stem cell transplantation (allo-SCT) for high-risk AML and allo-SCT for non-high-risk patients only in second complete remission after relapse. In the last decade, several drugs have been used in clinical trials to improve outcomes in pediatric AML treatment.
Collapse
|
6
|
Tong JTW, Harris PWR, Brimble MA, Kavianinia I. An Insight into FDA Approved Antibody-Drug Conjugates for Cancer Therapy. Molecules 2021; 26:5847. [PMID: 34641391 PMCID: PMC8510272 DOI: 10.3390/molecules26195847] [Citation(s) in RCA: 185] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 12/12/2022] Open
Abstract
The large number of emerging antibody-drug conjugates (ADCs) for cancer therapy has resulted in a significant market 'boom', garnering worldwide attention. Despite ADCs presenting huge challenges to researchers, particularly regarding the identification of a suitable combination of antibody, linker, and payload, as of September 2021, 11 ADCs have been granted FDA approval, with eight of these approved since 2017 alone. Optimism for this therapeutic approach is clear, despite the COVID-19 pandemic, 2020 was a landmark year for deals and partnerships in the ADC arena, suggesting that there remains significant interest from Big Pharma. Herein we review the enthusiasm for ADCs by focusing on the features of those approved by the FDA, and offer some thoughts as to where the field is headed.
Collapse
Affiliation(s)
- Juliana T. W. Tong
- School of Chemical Sciences, The University of Auckland, Auckland 1010, New Zealand; (J.T.W.T.); (P.W.R.H.)
- Maurice Wilkins Centre for Molecular Biodiversity, The University of Auckland, Auckland 1010, New Zealand
| | - Paul W. R. Harris
- School of Chemical Sciences, The University of Auckland, Auckland 1010, New Zealand; (J.T.W.T.); (P.W.R.H.)
- Maurice Wilkins Centre for Molecular Biodiversity, The University of Auckland, Auckland 1010, New Zealand
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
| | - Margaret A. Brimble
- School of Chemical Sciences, The University of Auckland, Auckland 1010, New Zealand; (J.T.W.T.); (P.W.R.H.)
- Maurice Wilkins Centre for Molecular Biodiversity, The University of Auckland, Auckland 1010, New Zealand
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
| | - Iman Kavianinia
- School of Chemical Sciences, The University of Auckland, Auckland 1010, New Zealand; (J.T.W.T.); (P.W.R.H.)
- Maurice Wilkins Centre for Molecular Biodiversity, The University of Auckland, Auckland 1010, New Zealand
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
| |
Collapse
|
7
|
Buongervino S, Lane MV, Garrigan E, Zhelev DV, Dimitrov DS, Bosse KR. Antibody-Drug Conjugate Efficacy in Neuroblastoma: Role of Payload, Resistance Mechanisms, Target Density, and Antibody Internalization. Mol Cancer Ther 2021; 20:2228-2239. [PMID: 34465595 DOI: 10.1158/1535-7163.mct-20-1034] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 05/18/2021] [Accepted: 08/20/2021] [Indexed: 11/16/2022]
Abstract
Antibody-drug conjugates (ADC) are a targeted cancer therapy that utilize the specificity of antibodies to deliver potent drugs selectively to tumors. Here we define the complex interaction among factors that dictate ADC efficacy in neuroblastoma by testing both a comprehensive panel of ADC payloads in a diverse set of neuroblastoma cell lines and utilizing the glypican 2 (GPC2)-targeting D3-GPC2-PBD ADC to study the role of target antigen density and antibody internalization in ADC efficacy in neuroblastoma. We first find that DNA binding drugs are significantly more cytotoxic to neuroblastomas than payloads that bind tubulin or inhibit DNA topoisomerase 1. We additionally show that neuroblastomas with high expression of the ABCB1 drug transporter or that harbor a TP53 mutation are significantly more resistant to tubulin and DNA/DNA topoisomerase 1 binding payloads, respectively. Next, we utilized the GPC2-specific D3-GPC2-IgG1 antibody to show that neuroblastomas internalize this antibody/GPC2 complex at significantly different rates and that these antibody internalization kinetics correlate significantly with GPC2 cell surface density. However, sensitivity to pyrrolobenzodiazepine (PBD) dimers primarily dictated sensitivity to the corresponding D3-GPC2-PBD ADC, overall having a larger influence on ADC efficacy than GPC2 cell surface density or antibody internalization. Finally, we utilized GPC2 isogenic Kelly neuroblastoma cells with different levels of cell surface GPC2 expression to define the threshold of target density required for ADC efficacy. Taken together, DNA binding ADC payloads should be prioritized for development for neuroblastoma given their superior efficacy and considering that ADC payload sensitivity is a major determinant of ADC efficacy.
Collapse
Affiliation(s)
- Samantha Buongervino
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia; Philadelphia, Pennsylvania
| | - Maria V Lane
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia; Philadelphia, Pennsylvania
| | - Emily Garrigan
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia; Philadelphia, Pennsylvania
| | - Doncho V Zhelev
- Department of Medicine, University of Pittsburgh School of Medicine; Pittsburgh Pennsylvania
| | - Dimiter S Dimitrov
- Department of Medicine, University of Pittsburgh School of Medicine; Pittsburgh Pennsylvania
| | - Kristopher R Bosse
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia; Philadelphia, Pennsylvania. .,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
8
|
Liu Y, Wei J, Liu J, Ma W, Duan Y, Liu D. Novel AXL-targeted agents overcome FLT3 inhibitor resistance in FLT3-ITD + acute myeloid leukemia cells. Oncol Lett 2021; 21:397. [PMID: 33777220 PMCID: PMC7988696 DOI: 10.3892/ol.2021.12658] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 02/10/2021] [Indexed: 01/02/2023] Open
Abstract
AXL receptor tyrosine kinase (AXL) upregulation mediates drug resistance in several types of human cancer and has become a therapeutic target worthy of exploration. The present study investigated AXL antigen expression and the effects of novel AXL-targeted agents in acute myeloid leukemia (AML) cells. AXL antigen expression in drug-sensitive and drug-resistant human AML cell lines, and AML blast cells from 57 patients with different clinical characteristics, was analyzed by flow cytometry and compared. Furthermore, the effects of the novel AXL antibody DAXL-88, antibody-drug conjugate DAXL-88-monomethyl auristatin E (MMAE), AXL small molecule inhibitor R428 and their combination with FMS-like tyrosine kinase 3 (FLT3) inhibitor quizartinib (AC220) in AML cells were analyzed by Cell Counting Kit-8 assay, flow cytometry and western blotting. The present study revealed that AXL antigen expression was upregulated in FLT3-internal tandem duplication (ITD)/tyrosine kinase domain mutation-positive (TKD)+ AML blast cells compared with FLT3-ITD/TKD- AML cells. Additionally, AXL antigen expression was markedly upregulated in the AC220-resistant FLT3-ITD+ MV4-11 cell line (MV4-11/AC220) and in FLT3 inhibitor-resistant blast cells from a patient with FLT3-ITD+ AML compared with parental sensitive cells. The AXL-targeted agents DAXL-88, DAXL-88-MMAE and R428 exhibited dose-dependent cytotoxic effects on FLT3-mutant AML cell lines (THP-1, MV4-11 and MV4-11/AC220) and blast cells from patients with FLT3-ITD+ AML. Combinations of AXL-targeted agents with AC220 exerted synergistic cytotoxic effects and induced apoptosis in MV4-11/AC220 cells and FLT3 inhibitor-resistant blast cells. The antileukemic effect of DAXL-88 and DAXL-88-MMAE may rely on their ability to block AXL, FLT3 and their downstream signaling pathways. The present study demonstrated the association between AXL antigen expression upregulation and drug resistance in FLT3-ITD+ AML, and proposed a method for overcoming FLT3 inhibitor resistance of FLT3-ITD+ AML using novel AXL-targeted agents.
Collapse
Affiliation(s)
- Yi Liu
- Department of Hematology, Chinese PLA Medical School, Beijing 100853, P.R. China.,Department of Hematology, The Sixth Medical Center of PLA General Hospital, Beijing 100048, P.R. China
| | - Jing Wei
- Department of Hematology, The Sixth Medical Center of PLA General Hospital, Beijing 100048, P.R. China
| | - Jiaxin Liu
- Department of Hematology, The Sixth Medical Center of PLA General Hospital, Beijing 100048, P.R. China
| | - Weina Ma
- Department of Hematology, The Sixth Medical Center of PLA General Hospital, Beijing 100048, P.R. China
| | - Yanting Duan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P.R. China.,Beijing Key Laboratory of Therapeutic Gene Engineering Antibody, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P.R. China
| | - Daihong Liu
- Department of Hematology, Chinese PLA Medical School, Beijing 100853, P.R. China.,Department of Hematology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| |
Collapse
|
9
|
Han YC, Kahler J, Piché-Nicholas N, Hu W, Thibault S, Jiang F, Leal M, Katragadda M, Maderna A, Dushin R, Prashad N, Charati MB, Clark T, Tumey LN, Tan X, Giannakou A, Rosfjord E, Gerber HP, Tchistiakova L, Loganzo F, O'Donnell CJ, Sapra P. Development of Highly Optimized Antibody-Drug Conjugates against CD33 and CD123 for Acute Myeloid Leukemia. Clin Cancer Res 2021; 27:622-631. [PMID: 33148666 DOI: 10.1158/1078-0432.ccr-20-2149] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/15/2020] [Accepted: 10/29/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Mortality due to acute myeloid leukemia (AML) remains high, and the management of relapsed or refractory AML continues to be therapeutically challenging. The reapproval of Mylotarg, an anti-CD33-calicheamicin antibody-drug conjugate (ADC), has provided a proof of concept for an ADC-based therapeutic for AML. Several other ADCs have since entered clinical development of AML, but have met with limited success. We sought to develop a next-generation ADC for AML with a wide therapeutic index (TI) that overcomes the shortcomings of previous generations of ADCs. EXPERIMENTAL DESIGN We compared the TI of our novel CD33-targeted ADC platform with other currently available CD33-targeted ADCs in preclinical models of AML. Next, using this next-generation ADC platform, we performed a head-to-head comparison of two attractive AML antigens, CD33 and CD123. RESULTS Our novel ADC platform offered improved safety and TI when compared with certain currently available ADC platforms in preclinical models of AML. Differentiation between the CD33- and CD123-targeted ADCs was observed in safety studies conducted in cynomolgus monkeys. The CD33-targeted ADC produced severe hematologic toxicity, whereas minimal hematologic toxicity was observed with the CD123-targeted ADC at the same doses and exposures. The improved toxicity profile of an ADC targeting CD123 over CD33 was consistent with the more restricted expression of CD123 in normal tissues. CONCLUSIONS We optimized all components of ADC design (i.e., leukemia antigen, antibody, and linker-payload) to develop an ADC that has the potential to translate into an effective new therapy against AML.
Collapse
Affiliation(s)
- Yoon-Chi Han
- Pfizer Inc., Oncology Research & Development, Pearl River, New York.
| | - Jennifer Kahler
- Pfizer Inc., Oncology Research & Development, Pearl River, New York
| | | | - Wenyue Hu
- Pfizer Inc., Drug Safety Research & Development, La Jolla, California
| | - Stephane Thibault
- Pfizer Inc., Drug Safety Research & Development, La Jolla, California
| | - Fan Jiang
- Pfizer Inc., Oncology Research & Development, Pearl River, New York
| | - Mauricio Leal
- Pfizer Inc., BioMedicine Design, Cambridge, Massachusetts and Pearl River, New York
| | - Madan Katragadda
- Pfizer Inc., BioMedicine Design, Cambridge, Massachusetts and Pearl River, New York
| | - Andreas Maderna
- Pfizer Inc., Worldwide Medicinal Chemistry, Groton, Connecticut
| | - Russell Dushin
- Pfizer Inc., Worldwide Medicinal Chemistry, Groton, Connecticut
| | - Nadira Prashad
- Pfizer Inc., Oncology Research & Development, Pearl River, New York
| | - Manoj B Charati
- Pfizer Inc., Oncology Research & Development, Pearl River, New York
| | | | - L Nathan Tumey
- Pfizer Inc., Worldwide Medicinal Chemistry, Groton, Connecticut
| | - Xingzhi Tan
- Pfizer Inc., Oncology Research & Development, Pearl River, New York
| | | | - Edward Rosfjord
- Pfizer Inc., Oncology Research & Development, Pearl River, New York
| | | | | | - Frank Loganzo
- Pfizer Inc., Oncology Research & Development, Pearl River, New York
| | | | - Puja Sapra
- Pfizer Inc., Oncology Research & Development, Pearl River, New York.
| |
Collapse
|
10
|
Conneely SE, Stevens AM. Acute Myeloid Leukemia in Children: Emerging Paradigms in Genetics and New Approaches to Therapy. Curr Oncol Rep 2021; 23:16. [PMID: 33439382 PMCID: PMC7806552 DOI: 10.1007/s11912-020-01009-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Acute myeloid leukemia (AML) in children remains a challenging disease to cure with suboptimal outcomes particularly when compared to the more common lymphoid leukemias. Recent advances in the genetic characterization of AML have enhanced understanding of individualized patient risk, which has also led to the development of new therapeutic strategies. Here, we review key cytogenetic and molecular features of pediatric AML and how new therapies are being used to improve outcomes. RECENT FINDINGS Recent studies have revealed an increasing number of mutations, including WT1, CBFA2T3-GLIS2, and KAT6A fusions, DEK-NUP214 and NUP98 fusions, and specific KMT2A rearrangements, which are associated with poor outcomes. However, outcomes are starting to improve with the addition of therapies such as gemtuzumab ozogamicin and FLT3 inhibitors, initially developed in adult AML. The combination of advanced risk stratification and ongoing improvements and innovations in treatment strategy will undoubtedly lead to better outcomes for children with AML.
Collapse
Affiliation(s)
- Shannon E Conneely
- Department of Pediatric Hematology/Oncology, Baylor College of Medicine/Texas Children's Hospital, 6701 Fannin, Suite 1510, Houston, TX, 77030, USA.
| | - Alexandra M Stevens
- Department of Pediatric Hematology/Oncology, Baylor College of Medicine/Texas Children's Hospital, 6701 Fannin, Suite 1510, Houston, TX, 77030, USA
| |
Collapse
|
11
|
Dean AQ, Luo S, Twomey JD, Zhang B. Targeting cancer with antibody-drug conjugates: Promises and challenges. MAbs 2021; 13:1951427. [PMID: 34291723 PMCID: PMC8300931 DOI: 10.1080/19420862.2021.1951427] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 06/29/2021] [Accepted: 06/29/2021] [Indexed: 01/03/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are a rapidly expanding class of biotherapeutics that utilize antibodies to selectively deliver cytotoxic drugs to the tumor site. As of May 2021, the U.S. Food and Drug Administration (FDA) has approved ten ADCs, namely Adcetris®, Kadcyla®, Besponsa®, Mylotarg®, Polivy®, Padcev®, Enhertu®, Trodelvy®, Blenrep®, and Zynlonta™ as monotherapy or combinational therapy for breast cancer, urothelial cancer, myeloma, acute leukemia, and lymphoma. In addition, over 80 investigational ADCs are currently being evaluated in approximately 150 active clinical trials. Despite the growing interest in ADCs, challenges remain to expand their therapeutic index (with greater efficacy and less toxicity). Recent advances in the manufacturing technology for the antibody, payload, and linker combined with new bioconjugation platforms and state-of-the-art analytical techniques are helping to shape the future development of ADCs. This review highlights the current status of marketed ADCs and those under clinical investigation with a focus on translational strategies to improve product quality, safety, and efficacy.
Collapse
Affiliation(s)
- Alexis Q. Dean
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Shen Luo
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Julianne D. Twomey
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Baolin Zhang
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
12
|
Ali I, Mukhtar SD, Ali HS, Scotti MT, Scotti L. Advances in Nanoparticles as Anticancer Drug Delivery Vector: Need of this Century. Curr Pharm Des 2020; 26:1637-1649. [DOI: 10.2174/1381612826666200203124330] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 12/02/2019] [Indexed: 12/17/2022]
Abstract
Background:
Nanotechnology has contributed a great deal to the field of medical science. Smart drugdelivery
vectors, combined with stimuli-based characteristics, are becoming increasingly important. The use of
external and internal stimulating factors can have enormous benefits and increase the targeting efficiency of
nanotechnology platforms. The pH values of tumor vascular tissues are acidic in nature, allowing the improved
targeting of anticancer drug payloads using drug-delivery vectors. Nanopolymers are smart drug-delivery vectors
that have recently been developed and recommended for use by scientists because of their potential targeting
capabilities, non-toxicity and biocompatibility, and make them ideal nanocarriers for personalized drug delivery.
Method:
The present review article provides an overview of current advances in the use of nanoparticles (NPs) as
anticancer drug-delivery vectors.
Results:
This article reviews the molecular basis for the use of NPs in medicine, including personalized medicine,
personalized therapy, emerging vistas in anticancer therapy, nanopolymer targeting, passive and active targeting
transports, pH-responsive drug carriers, biological barriers, computer-aided drug design, future challenges and
perspectives, biodegradability and safety.
Conclusions:
This article will benefit academia, researchers, clinicians, and government authorities by providing a
basis for further research advancements.
Collapse
Affiliation(s)
- Imran Ali
- Department of Chemistry, College of Sciences, Taibah University, Al-Medina Al-Munawara – 41477, Saudi Arabia
| | - Sofi D. Mukhtar
- Department of Chemistry, Jamia Millia Islamia (Central University) New Delhi-110025, India
| | - Heyam S. Ali
- Department of Pharmaceutics, University of Khartoum, Khartoum, Sudan
| | - Marcus T. Scotti
- Cheminformatics Laboratory- Postgraduate Program in Natural Products and Synthetic Bioactive, Federal University of Paraíba-Campus I 58051-970, João Pessoa, PB, Brazil
| | - Luciana Scotti
- Teaching and Research Management - University Hospital, Cheminformatics Laboratory- Postgraduate Program in Natural Products and Synthetic Bioactive, Federal University of Paraíba-Campus I, 58051-970, João Pessoa, PB, Brazil
| |
Collapse
|
13
|
Wang X, Hu Y, Mo J, Zhang J, Wang Z, Wei W, Li H, Xu Y, Ma J, Zhao J, Jin Z, Guo Z. Arsenene: A Potential Therapeutic Agent for Acute Promyelocytic Leukaemia Cells by Acting on Nuclear Proteins. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201913675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Xiuxiu Wang
- State Key Laboratory of Coordination ChemistryKey Laboratory of Mesoscopic Chemistry of MOEJiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center (ChemBIC)School of Chemistry and Chemical EngineeringNanjing University Nanjing 210023 China
| | - Yi Hu
- State Key Laboratory of Coordination ChemistryKey Laboratory of Mesoscopic Chemistry of MOEJiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center (ChemBIC)School of Chemistry and Chemical EngineeringNanjing University Nanjing 210023 China
| | - Jianbin Mo
- State Key Laboratory of Coordination ChemistryKey Laboratory of Mesoscopic Chemistry of MOEJiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center (ChemBIC)School of Chemistry and Chemical EngineeringNanjing University Nanjing 210023 China
| | - Jingyi Zhang
- State Key Laboratory of Coordination ChemistryKey Laboratory of Mesoscopic Chemistry of MOEJiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center (ChemBIC)School of Chemistry and Chemical EngineeringNanjing University Nanjing 210023 China
| | - Zhenzhen Wang
- Medical school of Nanjing UniversityNanjing University Nanjing 210023 China
| | - Wei Wei
- School of Life SciencesNanjing University Nanjing 210023 China
| | - Huanlin Li
- School of Life SciencesNanjing University Nanjing 210023 China
| | - Yun Xu
- School of Life SciencesNanjing University Nanjing 210023 China
| | - Jing Ma
- State Key Laboratory of Coordination ChemistryKey Laboratory of Mesoscopic Chemistry of MOEJiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center (ChemBIC)School of Chemistry and Chemical EngineeringNanjing University Nanjing 210023 China
| | - Jing Zhao
- State Key Laboratory of Coordination ChemistryKey Laboratory of Mesoscopic Chemistry of MOEJiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center (ChemBIC)School of Chemistry and Chemical EngineeringNanjing University Nanjing 210023 China
| | - Zhong Jin
- State Key Laboratory of Coordination ChemistryKey Laboratory of Mesoscopic Chemistry of MOEJiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center (ChemBIC)School of Chemistry and Chemical EngineeringNanjing University Nanjing 210023 China
| | - Zijian Guo
- State Key Laboratory of Coordination ChemistryKey Laboratory of Mesoscopic Chemistry of MOEJiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center (ChemBIC)School of Chemistry and Chemical EngineeringNanjing University Nanjing 210023 China
| |
Collapse
|
14
|
Wang X, Hu Y, Mo J, Zhang J, Wang Z, Wei W, Li H, Xu Y, Ma J, Zhao J, Jin Z, Guo Z. Arsenene: A Potential Therapeutic Agent for Acute Promyelocytic Leukaemia Cells by Acting on Nuclear Proteins. Angew Chem Int Ed Engl 2020; 59:5151-5158. [PMID: 31891659 DOI: 10.1002/anie.201913675] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Indexed: 01/01/2023]
Abstract
Arsenene has recently emerged as a promising new two-dimensional material for biomedical applications because of its excellent optical and electronic properties. Herein, novel 2D arsenene nanosheets were synthesized and shown to be effective against NB4 promyelocytic leukaemia (APL) cells (82 % inhibition) as well as inducing apoptosis while showing no toxicity towards normal cells. The high zeta potential, small size, and the planar structure were crucial to the toxicity of the materials. Label-free proteomic profiling analysis suggested that arsenene affected nuclear DNA replication, nucleotide excision repair, and pyrimidine metabolism pathways by downregulating the DNA polymerases POLE, POLD1, POLD2, and POLD3. Mass spectrometric studies showed that arsenene bound mainly to nuclear nucleotide acid binding proteins in NB4 cells and further cellular fluorescence studies revealed that the arsenene destroyed the nuclei. In vivo toxicity tests in mice also indicated the physiological biosafety of arsenene.
Collapse
Affiliation(s)
- Xiuxiu Wang
- State Key Laboratory of Coordination Chemistry, Key Laboratory of Mesoscopic Chemistry of MOE, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yi Hu
- State Key Laboratory of Coordination Chemistry, Key Laboratory of Mesoscopic Chemistry of MOE, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Jianbin Mo
- State Key Laboratory of Coordination Chemistry, Key Laboratory of Mesoscopic Chemistry of MOE, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Jingyi Zhang
- State Key Laboratory of Coordination Chemistry, Key Laboratory of Mesoscopic Chemistry of MOE, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Zhenzhen Wang
- Medical school of Nanjing University, Nanjing University, Nanjing, 210023, China
| | - Wei Wei
- School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Huanlin Li
- School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Yun Xu
- School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Jing Ma
- State Key Laboratory of Coordination Chemistry, Key Laboratory of Mesoscopic Chemistry of MOE, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Jing Zhao
- State Key Laboratory of Coordination Chemistry, Key Laboratory of Mesoscopic Chemistry of MOE, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Zhong Jin
- State Key Laboratory of Coordination Chemistry, Key Laboratory of Mesoscopic Chemistry of MOE, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry, Key Laboratory of Mesoscopic Chemistry of MOE, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| |
Collapse
|
15
|
Sarmento-Ribeiro AB, Scorilas A, Gonçalves AC, Efferth T, Trougakos IP. The emergence of drug resistance to targeted cancer therapies: Clinical evidence. Drug Resist Updat 2019; 47:100646. [PMID: 31733611 DOI: 10.1016/j.drup.2019.100646] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 12/14/2022]
Abstract
For many decades classical anti-tumor therapies included chemotherapy, radiation and surgery; however, in the last two decades, following the identification of the genomic drivers and main hallmarks of cancer, the introduction of therapies that target specific tumor-promoting oncogenic or non-oncogenic pathways, has revolutionized cancer therapeutics. Despite the significant progress in cancer therapy, clinical oncologists are often facing the primary impediment of anticancer drug resistance, as many cancer patients display either intrinsic chemoresistance from the very beginning of the therapy or after initial responses and upon repeated drug treatment cycles, acquired drug resistance develops and thus relapse emerges, resulting in increased mortality. Our attempts to understand the molecular basis underlying these drug resistance phenotypes in pre-clinical models and patient specimens revealed the extreme plasticity and adaptive pathways employed by tumor cells, being under sustained stress and extensive genomic/proteomic instability due to the applied therapeutic regimens. Subsequent efforts have yielded more effective inhibitors and combinatorial approaches (e.g. the use of specific pharmacologic inhibitors with immunotherapy) that exhibit synergistic effects against tumor cells, hence enhancing therapeutic indices. Furthermore, new advanced methodologies that allow for the early detection of genetic/epigenetic alterations that lead to drug chemoresistance and prospective validation of biomarkers which identify patients that will benefit from certain drug classes, have started to improve the clinical outcome. This review discusses emerging principles of drug resistance to cancer therapies targeting a wide array of oncogenic kinases, along with hedgehog pathway and the proteasome and apoptotic inducers, as well as epigenetic and metabolic modulators. We further discuss mechanisms of resistance to monoclonal antibodies, immunomodulators and immune checkpoint inhibitors, potential biomarkers of drug response/drug resistance, along with possible new therapeutic avenues for the clinicians to combat devastating drug resistant malignancies. It is foreseen that these topics will be major areas of focused multidisciplinary translational research in the years to come.
Collapse
Affiliation(s)
- Ana Bela Sarmento-Ribeiro
- Laboratory of Oncobiology and Hematology and University Clinic of Hematology and Coimbra Institute for Clinical and Biomedical Research - Group of Environment Genetics and Oncobiology (iCBR/CIMAGO), Faculty of Medicine, University of Coimbra (FMUC), Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Hematology Department, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal.
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Ana Cristina Gonçalves
- Laboratory of Oncobiology and Hematology and University Clinic of Hematology and Coimbra Institute for Clinical and Biomedical Research - Group of Environment Genetics and Oncobiology (iCBR/CIMAGO), Faculty of Medicine, University of Coimbra (FMUC), Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Ioannis P Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Greece.
| |
Collapse
|
16
|
Strategies for Targeting Cancer Immunotherapy Through Modulation of the Tumor Microenvironment. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2019. [DOI: 10.1007/s40883-019-00113-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
17
|
Yaghoubi S, Karimi MH, Lotfinia M, Gharibi T, Mahi-Birjand M, Kavi E, Hosseini F, Sineh Sepehr K, Khatami M, Bagheri N, Abdollahpour-Alitappeh M. Potential drugs used in the antibody-drug conjugate (ADC) architecture for cancer therapy. J Cell Physiol 2019; 235:31-64. [PMID: 31215038 DOI: 10.1002/jcp.28967] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 05/20/2019] [Indexed: 01/04/2023]
Abstract
Cytotoxic small-molecule drugs have a major influence on the fate of antibody-drug conjugates (ADCs). An ideal cytotoxic agent should be highly potent, remain stable while linked to ADCs, kill the targeted tumor cell upon internalization and release from the ADCs, and maintain its activity in multidrug-resistant tumor cells. Lessons learned from successful and failed experiences in ADC development resulted in remarkable progress in the discovery and development of novel highly potent small molecules. A better understanding of such small-molecule drugs is important for development of effective ADCs. The present review discusses requirements making a payload appropriate for antitumor ADCs and focuses on the main characteristics of commonly-used cytotoxic payloads that showed acceptable results in clinical trials. In addition, the present study represents emerging trends and recent advances of payloads used in ADCs currently under clinical trials.
Collapse
Affiliation(s)
- Sajad Yaghoubi
- Department of Clinical Microbiology, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | | | - Majid Lotfinia
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran.,Core Research Lab, Kashan University of Medical Sciences, Kashan, Iran
| | - Tohid Gharibi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Motahare Mahi-Birjand
- Infectious Disease Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Esmaeil Kavi
- Department of Nursing, School of Nursing, Larestan University of Medical Sciences, Larestan, Iran
| | - Fahimeh Hosseini
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Koushan Sineh Sepehr
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehrdad Khatami
- NanoBioelectrochemistry Research Center, Bam University of Medical Sciences, Bam, Iran
| | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | |
Collapse
|
18
|
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
|
19
|
Kunos CA, Capala J, Ivy SP. Radiopharmaceuticals for Relapsed or Refractory Leukemias. Front Oncol 2019; 9:97. [PMID: 30859091 PMCID: PMC6397856 DOI: 10.3389/fonc.2019.00097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 02/04/2019] [Indexed: 12/21/2022] Open
Abstract
Radiopharmaceuticals, meaning drugs that hold a radionuclide intended for use in cancer patients for treatment of their disease or for palliation of their disease-related symptoms, have gained new interest for clinical development in adult patients with relapsed or refractory leukemia. About one-third of adult patients outlive their leukemia, with the remainder unable to attain complete remission status following the first phase of treatment due to refractory bone marrow or blood residual microscopic disease. The National Cancer Institute (NCI) Cancer Therapy Evaluation Program conducted 49 phase 1-1b trials in adult patients with leukemia between 1986 and 2017 in an effort to discover tolerated and effective therapeutic drug combinations intended to improve remission and mortality rates. None of these trials involved radiopharmaceuticals. In this article, the NCI perspective on the challenges encountered in and on the future potential of radiopharmaceuticals alone or in combination for adult patients with relapsed or refractory leukemia is discussed. An effort is underway already to build-up the NCI's clinical trial enterprise infrastructure for radiopharmaceutical clinical development.
Collapse
Affiliation(s)
- Charles A Kunos
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, United States
| | - Jacek Capala
- Radiation Research Program, National Cancer Institute, Bethesda, MD, United States
| | - Susan Percy Ivy
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, United States
| |
Collapse
|
20
|
Kovtun Y, Noordhuis P, Whiteman KR, Watkins K, Jones GE, Harvey L, Lai KC, Portwood S, Adams S, Sloss CM, Schuurhuis GJ, Ossenkoppele G, Wang ES, Pinkas J. IMGN779, a Novel CD33-Targeting Antibody-Drug Conjugate with DNA-Alkylating Activity, Exhibits Potent Antitumor Activity in Models of AML. Mol Cancer Ther 2018; 17:1271-1279. [PMID: 29588393 DOI: 10.1158/1535-7163.mct-17-1077] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/18/2018] [Accepted: 03/23/2018] [Indexed: 11/16/2022]
Abstract
The myeloid differentiation antigen CD33 has long been exploited as a target for antibody-based therapeutic approaches in acute myeloid leukemia (AML). Validation of this strategy was provided with the approval of the CD33-targeting antibody-drug conjugate (ADC) gemtuzumab ozogamicin in 2000; the clinical utility of this agent, however, has been hampered by safety concerns. Thus, the full potential of CD33-directed therapy in AML remains to be realized, and considerable interest exists in the design and development of more effective ADCs that confer high therapeutic indices and favorable tolerability profiles. Here, we describe the preclinical characterization of a novel CD33-targeting ADC, IMGN779, which utilizes a unique DNA-alkylating payload to achieve potent antitumor effects with good tolerability. The payload, DGN462, is prototypical of a novel class of purpose-created indolinobenzodiazeprine pseudodimers, termed IGNs. With low picomolar potency, IMGN779 reduced viability in a panel of AML cell lines in vitro Mechanistically, the cytotoxic activity of IMGN779 involved DNA damage, cell-cycle arrest, and apoptosis consistent with the mode of action of DGN462. Moreover, IMGN779 was highly active against patient-derived AML cells, including those with adverse molecular abnormalities, and sensitivity correlated to CD33 expression levels. In vivo, IMGN779 displayed robust antitumor efficacy in multiple AML xenograft and disseminated disease models, as evidenced by durable tumor regressions and prolonged survival. Taken together, these findings identify IMGN779 as a promising new candidate for evaluation as a novel therapeutic in AML. Mol Cancer Ther; 17(6); 1271-9. ©2018 AACR.
Collapse
Affiliation(s)
| | - Paul Noordhuis
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | | | | | | | | | | | - Scott Portwood
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York
| | | | | | | | - Gert Ossenkoppele
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - Eunice S Wang
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York
| | | |
Collapse
|
21
|
Waghray D, Zhang Q. Inhibit or Evade Multidrug Resistance P-Glycoprotein in Cancer Treatment. J Med Chem 2017; 61:5108-5121. [PMID: 29251920 DOI: 10.1021/acs.jmedchem.7b01457] [Citation(s) in RCA: 280] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Multidrug resistance (MDR) is a major cause of failure in cancer chemotherapy. P-glycoprotein (P-gp), a promiscuous drug efflux pump, has been extensively studied for its association with MDR due to overexpression in cancer cells. Several P-gp inhibitors or modulators have been investigated in clinical trials in hope of circumventing MDR, with only limited success. Alternative strategies are actively pursued, such as the modification of existing drugs, development of new drugs, or combination of novel drug delivery agents to evade P-gp-dependent efflux. Despite the importance and numerous studies, these efforts have mostly been undertaken without a priori knowledge of how drugs interact with P-gp at the molecular level. This review highlights and discusses progress toward and challenges impeding drug development for inhibiting or evading P-gp in the context of our improved understanding of the structural basis and mechanism of P-gp-mediated MDR.
Collapse
Affiliation(s)
- Deepali Waghray
- Department of Integrative Structural and Computational Biology , The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Qinghai Zhang
- Department of Integrative Structural and Computational Biology , The Scripps Research Institute , La Jolla , California 92037 , United States
| |
Collapse
|
22
|
Kalim M, Chen J, Wang S, Lin C, Ullah S, Liang K, Ding Q, Chen S, Zhan J. Intracellular trafficking of new anticancer therapeutics: antibody-drug conjugates. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:2265-2276. [PMID: 28814834 PMCID: PMC5546728 DOI: 10.2147/dddt.s135571] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Antibody-drug conjugate (ADC) is a milestone in targeted cancer therapy that comprises of monoclonal antibodies chemically linked to cytotoxic drugs. Internalization of ADC takes place via clathrin-mediated endocytosis, caveolae-mediated endocytosis, and pinocytosis. Conjugation strategies, endocytosis and intracellular trafficking optimization, linkers, and drugs chemistry present a great challenge for researchers to eradicate tumor cells successfully. This inventiveness of endocytosis and intracellular trafficking has given considerable momentum recently to develop specific antibodies and ADCs to treat cancer cells. It is significantly advantageous to emphasize the endocytosis and intracellular trafficking pathways efficiently and to design potent engineered conjugates and biological entities to boost efficient therapies enormously for cancer treatment. Current studies illustrate endocytosis and intracellular trafficking of ADC, protein, and linker strategies in unloading and also concisely evaluate practically applicable ADCs.
Collapse
Affiliation(s)
- Muhammad Kalim
- Department of Biochemistry and Genetics, School of Medicine
| | - Jie Chen
- Department of Biochemistry and Genetics, School of Medicine
| | - Shenghao Wang
- Department of Biochemistry and Genetics, School of Medicine
| | - Caiyao Lin
- Department of Biochemistry and Genetics, School of Medicine
| | - Saif Ullah
- Department of Biochemistry and Genetics, School of Medicine
| | - Keying Liang
- Department of Biochemistry and Genetics, School of Medicine
| | - Qian Ding
- Department of Biochemistry and Genetics, School of Medicine
| | - Shuqing Chen
- Department of Pharmaceutical Analysis, College of Pharmaceutical Science, Zhejiang University, Hangzhou, People's Republic of China
| | - Jinbiao Zhan
- Department of Biochemistry and Genetics, School of Medicine
| |
Collapse
|
23
|
Stefan N, Gébleux R, Waldmeier L, Hell T, Escher M, Wolter FI, Grawunder U, Beerli RR. Highly Potent, Anthracycline-based Antibody–Drug Conjugates Generated by Enzymatic, Site-specific Conjugation. Mol Cancer Ther 2017; 16:879-892. [DOI: 10.1158/1535-7163.mct-16-0688] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 10/14/2016] [Accepted: 02/02/2017] [Indexed: 11/16/2022]
|
24
|
Enzyme-Based Strategies to Generate Site-Specifically Conjugated Antibody Drug Conjugates. NEXT GENERATION ANTIBODY DRUG CONJUGATES (ADCS) AND IMMUNOTOXINS 2017. [DOI: 10.1007/978-3-319-46877-8_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
25
|
Imrichova D, Messingerova L, Seres M, Kavcova H, Pavlikova L, Coculova M, Breier A, Sulova Z. Selection of resistant acute myeloid leukemia SKM-1 and MOLM-13 cells by vincristine-, mitoxantrone- and lenalidomide-induced upregulation of P-glycoprotein activity and downregulation of CD33 cell surface exposure. Eur J Pharm Sci 2015; 77:29-39. [PMID: 26002042 DOI: 10.1016/j.ejps.2015.05.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 05/17/2015] [Accepted: 05/18/2015] [Indexed: 01/08/2023]
Abstract
Bone marrow cells and peripheral blood mononuclear cells obtained from both acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) patients contain upregulated levels of cell surface antigen CD33 compared with healthy controls. This difference enables the use of humanized anti-CD33 antibody conjugated to cytotoxic agents for CD33 targeted immunotherapy. However, the expression of the membrane-bound drug transporter P-glycoprotein (P-gp) has been shown to be critical for resistance against the cytotoxicity of a humanized anti-CD33 antibody conjugated to maytansine-derivative DM4. The aim of the present study was to examine whether the expression of P-gp in AML cell lines is associated with changes in CD33 expression. For this purpose, we established drug resistant variants of SKM-1 and MOLM-13 AML cell lines via the selection of parental cells for resistance to vincristine, mitoxantrone and lenalidomide. All three substances induced a multidrug resistance (MDR) phenotype in SKM-1 cells associated with strong upregulation of P-gp and downregulation of CD33. However, in MOLM-13 cells, the upregulation of P-gp and downregulation of CD33 were present only in cells selected for resistance to vincristine and mitoxantrone but not lenalidomide. Inverse expression of P-gp and CD33 were observed in all resistant variants of SKM-1 and MOLM-13 cells. The MDR phenotype of resistant variants of SKM-1 and MOLM-13 cells was associated with alterations in apoptotic regulatory proteins and downregulation of the multidrug resistance associated protein 1 and breast cancer resistance protein.
Collapse
Affiliation(s)
- D Imrichova
- Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences, Vlarska 5, 833 34 Bratislava, Slovak Republic
| | - L Messingerova
- Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences, Vlarska 5, 833 34 Bratislava, Slovak Republic; Institute of Biochemistry, Nutrition and Health Protection, Faculty of Chemical and Food Technology, Slovak University of Technology, Radlinskeho 9, 812 37 Bratislava, Slovak Republic
| | - M Seres
- Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences, Vlarska 5, 833 34 Bratislava, Slovak Republic
| | - H Kavcova
- Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences, Vlarska 5, 833 34 Bratislava, Slovak Republic
| | - L Pavlikova
- Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences, Vlarska 5, 833 34 Bratislava, Slovak Republic
| | - M Coculova
- Institute of Biochemistry, Nutrition and Health Protection, Faculty of Chemical and Food Technology, Slovak University of Technology, Radlinskeho 9, 812 37 Bratislava, Slovak Republic
| | - A Breier
- Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences, Vlarska 5, 833 34 Bratislava, Slovak Republic; Institute of Biochemistry, Nutrition and Health Protection, Faculty of Chemical and Food Technology, Slovak University of Technology, Radlinskeho 9, 812 37 Bratislava, Slovak Republic.
| | - Z Sulova
- Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences, Vlarska 5, 833 34 Bratislava, Slovak Republic.
| |
Collapse
|
26
|
Verma VA, Pillow TH, DePalatis L, Li G, Phillips GL, Polson AG, Raab HE, Spencer S, Zheng B. The cryptophycins as potent payloads for antibody drug conjugates. Bioorg Med Chem Lett 2015; 25:864-8. [PMID: 25613677 DOI: 10.1016/j.bmcl.2014.12.070] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/17/2014] [Accepted: 12/19/2014] [Indexed: 01/30/2023]
Abstract
The cryptophycins are a potent class of cytotoxic agents that were evaluated as antibody drug conjugate (ADC) payloads. Free cryptophycin analog 1 displayed cell activity an order of magnitude more potent than approved ADC payloads MMAE and DM1. This potency increase was also reflected in the activity of the cryptophycin ADCs, attached via a either cleavable or non-cleavable linker.
Collapse
Affiliation(s)
- Vishal A Verma
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States.
| | - Thomas H Pillow
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Laura DePalatis
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Guangmin Li
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | | | - Andrew G Polson
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Helga E Raab
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Susan Spencer
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Bing Zheng
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| |
Collapse
|
27
|
|
28
|
Ji JA, Liu J, Wang YJ. Formulation Development for Antibody-Drug Conjugates. ANTIBODY-DRUG CONJUGATES 2015. [DOI: 10.1007/978-3-319-13081-1_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
29
|
Shefet-Carasso L, Benhar I. Antibody-targeted drugs and drug resistance--challenges and solutions. Drug Resist Updat 2014; 18:36-46. [PMID: 25476546 DOI: 10.1016/j.drup.2014.11.001] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 11/16/2014] [Accepted: 11/17/2014] [Indexed: 11/24/2022]
Abstract
Antibody-based therapy of various human malignancies has shown efficacy in the past 30 years and is now one of the most successful and leading strategies for targeted treatment of patients harboring hematological malignancies and solid tumors. Antibody-drug conjugates (ADCs) aim to take advantage of the affinity and specificity of monoclonal antibodies (mAbs) to selectively deliver potent cytotoxic drugs to antigen-expressing tumor cells. Key parameters for ADC include choosing the optimal components of the ADC (the antibody, the linker and the cytotoxic drug) and selecting the suitable cell-surface target antigen. Building on the success of recent FDA approval of brentuximab vedotin (Adcetris) and ado-trastuzumab emtansine (Kadcyla), ADCs are currently a class of drugs with a robust pipeline with clinical applications that are rapidly expanding. The more ADCs are being evaluated in preclinical models and clinical trials, the clearer are becoming the parameters and the challenges required for their therapeutic success. This rapidly growing knowledge and clinical experience are revealing novel modalities and mechanisms of resistance to ADCs, hence offering plausible solutions to such challenges. Here, we review the key parameters for designing a powerful ADC, focusing on how ADCs are addressing the challenge of multiple drug resistance (MDR) and its rational overcoming.
Collapse
Affiliation(s)
- LeeRon Shefet-Carasso
- Department of Molecular Microbiology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Ramat Aviv, Israel
| | - Itai Benhar
- Department of Molecular Microbiology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Ramat Aviv, Israel.
| |
Collapse
|
30
|
Han TH, Zhao B. Absorption, distribution, metabolism, and excretion considerations for the development of antibody-drug conjugates. Drug Metab Dispos 2014; 42:1914-20. [PMID: 25048520 DOI: 10.1124/dmd.114.058586] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are a class of therapeutics that are designed to deliver potent small-molecule drugs selectively to cells that express a specific target antigen while limiting systemic exposure to the drug. This is accomplished by conjugating a potent drug onto an antibody-based therapeutic with a linker that is exquisitely stable in plasma. The development of an effective ADC requires optimizing a number of design elements and an extensive understanding of absorption, distribution, metabolism/catabolism, and elimination (ADME) processes for the ADC construct. Furthermore, as ADCs are a combination of an antibody and small-molecule drug, understanding key aspects of the ADME of each individual component is needed. This review aims to provide considerations for the development of ADCs from an ADME point of view.
Collapse
Affiliation(s)
- Tae H Han
- Stem CentRx, Inc. (T.H.H.), South San Francisco, California; Seattle Genetics, Inc. (B.Z.), Bothell, Washington
| | - Baiteng Zhao
- Stem CentRx, Inc. (T.H.H.), South San Francisco, California; Seattle Genetics, Inc. (B.Z.), Bothell, Washington
| |
Collapse
|
31
|
Abstract
Immunoglobulins (Ig) or antibodies are heavy plasma proteins, with sugar chains added to amino-acid residues by N-linked glycosylation and occasionally by O-linked glycosylation. The versatility of antibodies is demonstrated by the various functions that they mediate such as neutralization, agglutination, fixation with activation of complement and activation of effector cells. Naturally occurring antibodies protect the organism against harmful pathogens, viruses and infections. In addition, almost any organic chemical induces antibody production of antibodies that would bind specifically to the chemical. These antibodies are often produced from multiple B cell clones and referred to as polyclonal antibodies. In recent years, scientists have exploited the highly evolved machinery of the immune system to produce structurally and functionally complex molecules such as antibodies from a single B clone, heralding the era of monoclonal antibodies. Most of the antibodies currently in the clinic, target components of the immune system, are not curative and seek to alleviate symptoms rather than cure disease. Our group used a novel strategy to identify reparative human monoclonal antibodies distinct from conventional antibodies. In this chapter, we discuss the therapeutic relevance of both polyclonal and monoclonal antibodies in clinic.
Collapse
Affiliation(s)
- Bharath Wootla
- Departments of Neurology and Immunology, Mayo Clinic, Rochester, MN, USA
| | | | | |
Collapse
|
32
|
Gasiorowski RE, Clark GJ, Bradstock K, Hart DNJ. Antibody therapy for acute myeloid leukaemia. Br J Haematol 2013; 164:481-95. [PMID: 24321020 DOI: 10.1111/bjh.12691] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Novel therapies with increased efficacy and decreased toxicity are desperately needed for the treatment of acute myeloid leukaemia (AML). The anti CD33 immunoconjugate, gemtuzumab ozogamicin (GO), was withdrawn with concerns over induction mortality and lack of efficacy. However a number of recent trials suggest that, particularly in AML with favourable cytogenetics, GO may improve overall survival. This data and the development of alternative novel monoclonal antibodies (mAb) have renewed interest in the area. Leukaemic stem cells (LSC) are identified as the subset of AML blasts that reproduces the leukaemic phenotype upon transplantation into immunosuppressed mice. AML relapse may be caused by chemoresistant LSC and this has refocused interest on identifying and targeting antigens specific for LSC. Several mAb have been developed that target LSC effectively in xenogeneic models but only a few have begun clinical evaluation. Antibody engineering may improve the activity of potential new therapeutics for AML. The encouraging results seen with bispecific T cell-engaging mAb-based molecules against CD19 in the treatment of B-cell acute lymphobalstic leukaemia, highlight the potential efficacy of engineered antibodies in the treatment of acute leukaemia. Potent engineered mAb, possibly targeting novel LSC antigens, offer hope for improving the current poor prognosis for AML.
Collapse
Affiliation(s)
- Robin E Gasiorowski
- ANZAC Research Institute, University of Sydney, Concord, NSW, Australia; Department of Haematology, Concord Cancer Centre, Concord Repatriation General Hospital, Concord, NSW, Australia
| | | | | | | |
Collapse
|
33
|
Cellular determinants for preclinical activity of a novel CD33/CD3 bispecific T-cell engager (BiTE) antibody, AMG 330, against human AML. Blood 2013; 123:554-61. [PMID: 24311721 DOI: 10.1182/blood-2013-09-527044] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
CD33 is a valid target for acute myeloid leukemia (AML) but has proven challenging for antibody-drug conjugates. Herein, we investigated the cellular determinants for the activity of the novel CD33/CD3-directed bispecific T-cell engager antibody, AMG 330. In the presence of T cells, AMG 330 was highly active against human AML cell lines and primary AML cells in a dose- and effector to target cell ratio-dependent manner. Using cell lines engineered to express wild-type CD33 at increased levels, we found a quantitative relationship between AMG 330 cytotoxicity and CD33 expression; in contrast, AMG 330 cytotoxicity was neither affected by common CD33 single nucleotide polymorphisms nor expression of the adenosine triphosphate-binding cassette (ABC) transporter proteins, P-glycoprotein or breast cancer resistance protein. Unlike bivalent CD33 antibodies, AMG 330 did not reduce surface CD33 expression. The epigenetic modifier drugs, panobinostat and azacitidine, increased CD33 expression in some cell lines and augmented AMG 330-induced cytotoxicity. These findings demonstrate that AMG 330 has potent CD33-dependent cytolytic activity in vitro, which can be further enhanced with other clinically available therapeutics. As it neither modulates CD33 expression nor is affected by ABC transporter activity, AMG 330 is highly promising for clinical exploration as it may overcome some limitations of previous CD33-targeted agents.
Collapse
|
34
|
Perez HL, Cardarelli PM, Deshpande S, Gangwar S, Schroeder GM, Vite GD, Borzilleri RM. Antibody-drug conjugates: current status and future directions. Drug Discov Today 2013; 19:869-81. [PMID: 24239727 DOI: 10.1016/j.drudis.2013.11.004] [Citation(s) in RCA: 336] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 09/27/2013] [Accepted: 11/04/2013] [Indexed: 01/25/2023]
Abstract
Antibody-drug conjugates (ADCs) aim to take advantage of the specificity of monoclonal antibodies (mAbs) to deliver potent cytotoxic drugs selectively to antigen-expressing tumor cells. Despite the simple concept, various parameters must be considered when designing optimal ADCs, such as selection of the appropriate antigen target and conjugation method. Each component of the ADC (the antibody, linker and drug) must also be optimized to fully realize the goal of a targeted therapy with improved efficacy and tolerability. Advancements over the past several decades have led to a new generation of ADCs comprising non-immunogenic mAbs, linkers with balanced stability and highly potent cytotoxic agents. Although challenges remain, recent clinical success has generated intense interest in this therapeutic class.
Collapse
Affiliation(s)
- Heidi L Perez
- Bristol-Myers Squibb Research & Development, Princeton, NJ 08543, USA
| | - Pina M Cardarelli
- Bristol-Myers Squibb Research & Development, Redwood City, CA 94063, USA
| | - Shrikant Deshpande
- Bristol-Myers Squibb Research & Development, Redwood City, CA 94063, USA
| | - Sanjeev Gangwar
- Bristol-Myers Squibb Research & Development, Redwood City, CA 94063, USA
| | | | - Gregory D Vite
- Bristol-Myers Squibb Research & Development, Princeton, NJ 08543, USA
| | | |
Collapse
|
35
|
SGN-CD33A: a novel CD33-targeting antibody-drug conjugate using a pyrrolobenzodiazepine dimer is active in models of drug-resistant AML. Blood 2013; 122:1455-63. [PMID: 23770776 DOI: 10.1182/blood-2013-03-491506] [Citation(s) in RCA: 306] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Outcomes in acute myeloid leukemia (AML) remain unsatisfactory, and novel treatments are urgently needed. One strategy explores antibodies and their drug conjugates, particularly those targeting CD33. Emerging data with gemtuzumab ozogamicin (GO) demonstrate target validity and activity in some patients with AML, but efficacy is limited by heterogeneous drug conjugation, linker instability, and a high incidence of multidrug resistance. We describe here the development of SGN-CD33A, a humanized anti-CD33 antibody with engineered cysteines conjugated to a highly potent, synthetic DNA cross-linking pyrrolobenzodiazepine dimer via a protease-cleavable linker. The use of engineered cysteine residues at the sites of drug linker attachment results in a drug loading of approximately 2 pyrrolobenzodiazepine dimers per antibody. In preclinical testing, SGN-CD33A is more potent than GO against a panel of AML cell lines and primary AML cells in vitro and in xenotransplantation studies in mice. Unlike GO, antileukemic activity is observed with SGN-CD33A in AML models with the multidrug-resistant phenotype. Mechanistic studies indicate that the cytotoxic effects of SGN-CD33A involve DNA damage with ensuing cell cycle arrest and apoptotic cell death. Together, these data suggest that SGN-CD33A has CD33-directed antitumor activity and support clinical testing of this novel therapeutic in patients with AML.
Collapse
|
36
|
Cowan AJ, Laszlo GS, Estey EH, Walter RB. Antibody-based therapy of acute myeloid leukemia with gemtuzumab ozogamicin. Front Biosci (Landmark Ed) 2013; 18:1311-34. [PMID: 23747885 DOI: 10.2741/4181] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Antibodies have created high expectations for effective yet tolerated therapeutics in acute myeloid leukemia (AML). Hitherto the most exploited target is CD33, a myeloid differentiation antigen found on AML blasts in most patients and, perhaps, leukemic stem cells in some. Treatment efforts have focused on conjugated antibodies, particularly gemtuzumab ozogamicin (GO), an anti-CD33 antibody carrying a toxic calicheamicin-g 1 derivative that, after intracellular hydrolytic release, induces DNA strand breaks, apoptosis, and cell death. Serving as paradigm for this strategy, GO was the first anti-cancer immunoconjugate to obtain regulatory approval in the U.S. While efficacious as monotherapy in acute promyelocytic leukemia (APL), GO alone induces remissions in less than 25-35% of non-APL AML patients. However, emerging data from well controlled trials now indicate that GO improves survival for many non-APL AML patients, supporting the conclusion that CD33 is a clinically relevant target for some disease subsets. It is thus unfortunate that GO has become unavailable in many parts of the world, and the drug's usefulness should be reconsidered and selected patients granted access to this immunoconjugate.
Collapse
Affiliation(s)
- Andrew J Cowan
- Hematology/Oncology Fellowship Program, University of Washington, Seattle, WA, USA
| | | | | | | |
Collapse
|
37
|
Takeshita A. Efficacy and resistance of gemtuzumab ozogamicin for acute myeloid leukemia. Int J Hematol 2013; 97:703-16. [PMID: 23709007 DOI: 10.1007/s12185-013-1365-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 05/07/2013] [Accepted: 05/08/2013] [Indexed: 11/24/2022]
Abstract
Seventy to 80 % of patients with acute myeloid leukemia (AML) achieve complete remission following intensive chemotherapy, but more than 50 % of patients in remission subsequently relapse, which is often associated with clinical drug resistance. Therapy based on monoclonal antibodies (mAbs) has been developed to increase the selectivity of cytotoxic agents by conjugating them with a mAb. Gemtuzumab ozogamicin (GO) is a conjugate of a cytotoxic agent, a calicheamicin derivative, linked to a recombinant humanized mAb directed against the CD33 antigen, which is expressed on leukemia cells from more than 90 % of patients with AML. This conjugated mAb was introduced following promising results from phase I and II studies. However, the initial phase III study did not confirm the efficacy of GO in combination with conventional chemotherapies. Several subsequent phase III studies have shown the efficacy of GO in favorable and intermediate risk AML. Several resistance mechanisms against GO have been reported. Multidrug resistant (MDR) P-glycoprotein (P-gp), a trans-membrane glycoprotein that pumps out many anti-leukemic agents from cells, also affects GO. For this reasons, GO has been used in combination with MDR modifiers, such as cyclosporine, and in cases without P-gp. Several investigators have reported successful results of the use of GO in acute promyelocytic leukemia (APL). GO has also been described as effective in cases relapsed after treatment with all-trans retinoic acid (ATRA), arsenic acid and conventional chemotherapeutic agents. The efficacy of GO will be studied mainly in a favorable risk of AML, such as core binding factor leukemia and APL. In addition, suitable combinations with other chemotherapies and administration schedules should be discussed.
Collapse
Affiliation(s)
- Akihiro Takeshita
- Transfusion and Cell Therapy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Japan.
| |
Collapse
|
38
|
Abstract
Toxin payloads, or drugs, are the crucial components of therapeutic antibody-drug conjugates (ADCs). This review will give an introduction on the requirements that make a toxic compound suitable to be used in an antitumoral ADC and will summarize the structural and mechanistic features of four drug families that yielded promising results in preclinical and clinical studies.
Collapse
Affiliation(s)
- Jan Anderl
- Heidelberg Pharma GmbH, Ladenburg, Germany
| | | | | | | |
Collapse
|
39
|
|
40
|
Abstract
Antibody conjugates are used in many therapeutic and research applications and are generated by chemically linking a cysteine or lysine residue to potent chemotherapeutic drugs or other functional groups through a flexible linker. Recently, we have engineered THIOMABs (antibodies with engineered reactive cysteine residues) for site-specific conjugation and showed that these antibody conjugates display homogeneous labeling with optimal in vitro and in vivo characteristics. Here, we describe protocols for engineering, selection, and site-specific conjugation of THIOMABs with thiol-reactive linkers.
Collapse
|
41
|
Salazar-Salinas K, Kubli-Garfias C, Seminario JM. Computational design of a CNT carrier for a high affinity bispecific anti-HER2 antibody based on trastuzumab and pertuzumab Fabs. J Mol Model 2012; 19:2797-810. [DOI: 10.1007/s00894-012-1638-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 10/07/2012] [Indexed: 12/25/2022]
|
42
|
Sultana S, Khan MR, Kumar M, Kumar S, Ali M. Nanoparticles-mediated drug delivery approaches for cancer targeting: a review. J Drug Target 2012; 21:107-25. [PMID: 22873288 DOI: 10.3109/1061186x.2012.712130] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cancer has become the leading cause of death among different populations of the world. The treatment is limited to chemotherapy, radiation, and surgery. Selective targeting to the tumor cells is possible by nanoparticles-based drug delivery system. It maximizes the drug concentration at the desired target and protects the surrounding healthy tissues at the same time. To improve the targeting potential of the anticancer drugs, nanoparticles were optimized for the size and surface characteristics to enhance their circulation time and targeting efficiency. Passive targeting involves surface modification with polyethylene glycol to avoid its elimination by natural body defense mechanism. Active targeting involves chemical interaction with certain antigen, receptors, and genes which are over expressed during progression of disease. In addition, the article highlights recent developments in "smart"-stimulus-responsive-drug carriers designed to enhance the localization and efficacy of therapeutic payloads as compared with free drug. Enhanced targeting potential, imaging, and controlled release of drugs or therapeutic molecules could be possible through multi-functional nanocarrier. Such multi-faceted, versatile nanocarriers and drug delivery systems promise a substantial increase in the efficacy of diagnostic and therapeutic applications in pharmaceutical sciences.
Collapse
|
43
|
Matsumoto T, Jimi S, Hara S, Takamatsu Y, Suzumiya J, Tamura K. Importance of inducible multidrug resistance 1 expression in HL-60 cells resistant to gemtuzumab ozogamicin. Leuk Lymphoma 2012; 53:1399-405. [DOI: 10.3109/10428194.2012.656102] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
44
|
Herrmann H, Cerny-Reiterer S, Gleixner KV, Blatt K, Herndlhofer S, Rabitsch W, Jäger E, Mitterbauer-Hohendanner G, Streubel B, Selzer E, Schwarzinger I, Sperr WR, Valent P. CD34(+)/CD38(-) stem cells in chronic myeloid leukemia express Siglec-3 (CD33) and are responsive to the CD33-targeting drug gemtuzumab/ozogamicin. Haematologica 2011; 97:219-26. [PMID: 21993666 DOI: 10.3324/haematol.2010.035006] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND CD33 is a well-known stem cell target in acute myeloid leukemia. So far, however, little is known about expression of CD33 on leukemic stem cells in chronic leukemias. DESIGN AND METHODS We analyzed expression of CD33 in leukemic progenitors in chronic myeloid leukemia by multi-color flow cytometry and quantitative polymerase chain reaction. In addition, the effects of a CD33-targeting drug, gemtuzumab/ozogamicin, were examined. RESULTS As assessed by flow cytometry, stem cell-enriched CD34(+)/CD38(-)/CD123(+) leukemic cells expressed significantly higher levels of CD33 compared to normal CD34(+)/CD38(-) stem cells. Moreover, highly enriched leukemic CD34(+)/CD38(-) cells (>98% purity) displayed higher levels of CD33 mRNA. In chronic phase patients, CD33 was found to be expressed invariably on most or all stem cells, whereas in accelerated or blast phase of the disease, the levels of CD33 on stem cells varied from donor to donor. The MDR1 antigen, supposedly involved in resistance against ozogamicin, was not detectable on leukemic CD34(+)/CD38(-) cells. Correspondingly, gemtuzumab/ozogamicin produced growth inhibition in leukemic progenitor cells in all patients tested. The effects of gemtuzumab/ozogamicin were dose-dependent, occurred at low concentrations, and were accompanied by apoptosis in suspension culture. Moreover, the drug was found to inhibit growth of leukemic cells in a colony assay and long-term culture-initiating cell assay. Finally, gemtuzumab/ozogamicin was found to synergize with nilotinib and bosutinib in inducing growth inhibition in leukemic cells. CONCLUSIONS CD33 is expressed abundantly on immature CD34(+)/CD38(-) stem cells and may serve as a stem cell target in chronic myeloid leukemia.
Collapse
|
45
|
Breccia M, Lo-Coco F. Gemtuzumab ozogamicin for the treatment of acute promyelocytic leukemia: mechanisms of action and resistance, safety and efficacy. Expert Opin Biol Ther 2010; 11:225-34. [PMID: 21142804 DOI: 10.1517/14712598.2011.543895] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Acute promyelocytic leukemia (APL) is characterized by peculiar biological features and high sensitivity to therapeutic agents such as anthracyclines, all-trans retinoic acid (ATRA) and arsenic trioxide (ATO). Because cure rates of up to 80 - 90% have been reported using various combinations of the above agents, future strategies will probably aim at reducing therapy-related toxicity while maintaining therapeutic efficacy. Gemtuzumab ozogamicin (GO) is a calicheamicin-conjugated mAb directed against CD33, a surface antigen highly expressed on APL blasts. GO has been shown to be effective in this disease and better tolerated than conventional chemotherapy. AREAS COVERED This review looks at the mechanism of action, pathways associated with resistance and toxicity profile of GO. Reported experience on the use of GO for relapsed or newly diagnosed APL is also discussed along with evidence on its efficacy and relative tolerability in APL management. In addition to its activity in advanced disease, data suggest that GO in various combinations may replace chemotherapy in APL front-line therapy. This should apply in particular to some subsets such as elderly patients or those unfit to receive conventional chemotherapy. EXPERT OPINION GO has proven effective and relatively safe as a single agent in advanced APL. In combinations with ATRA and/or ATO, GO may substitute for conventional chemotherapy of APL, particularly in unfit patients.
Collapse
Affiliation(s)
- Massimo Breccia
- Sapienza University, Department of Cellular Biotechnologies and Hematology, Via Benevento 6, 00161 Roma, Rome, Italy.
| | | |
Collapse
|
46
|
Junutula JR, Flagella KM, Graham RA, Parsons KL, Ha E, Raab H, Bhakta S, Nguyen T, Dugger DL, Li G, Mai E, Lewis Phillips GD, Hiraragi H, Fuji RN, Tibbitts J, Vandlen R, Spencer SD, Scheller RH, Polakis P, Sliwkowski MX. Engineered Thio-Trastuzumab-DM1 Conjugate with an Improved Therapeutic Index to Target Human Epidermal Growth Factor Receptor 2–Positive Breast Cancer. Clin Cancer Res 2010; 16:4769-78. [DOI: 10.1158/1078-0432.ccr-10-0987] [Citation(s) in RCA: 215] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
47
|
Abdool A, Yeh CH, Kantarjian H, O'Brien S, Bruey J, Giles F, Albitar M. Circulating CD33 and its clinical value in acute leukemia. Exp Hematol 2010; 38:462-71. [PMID: 20362641 DOI: 10.1016/j.exphem.2010.03.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2009] [Revised: 02/23/2010] [Accepted: 03/25/2010] [Indexed: 02/03/2023]
Abstract
OBJECTIVE CD33 is a cell surface antigen for committed myelomonocytic lineage. We explored the potential of detecting CD33 as cell-free circulating protein in patients with leukemia. MATERIALS AND METHODS We developed a quantitative bead-based immunoflow cytometry assay to measure cell-free circulating CD33 (cCD33) levels in the plasma of patients with acute leukemia, and correlated these results with corresponding clinical behavior. We measured cCD33 levels in the plasma of 48 healthy subjects and in patients with acute myelogenous leukemia (n = 98), acute lymphoblastic leukemia (n = 46), myelodysplastic syndrome (MDS) (n = 50), and myeloproliferative disorder (n = 49). RESULTS Patients with acute myeloid leukemia and myeloproliferative disorders had significantly higher concentrations of cCD33 than the other patient groups and normal individuals (p = 0.0001), and among these groups, MDS patients displayed the lowest cCD33 levels (p = 0.02). Circulating CD33 values correlated positively with the CD33(+) blast cell counts in these patients. While there was no correlation between cCD33 levels and survival in acute myelogenous leukemia and MDS, higher cCD33 plasma concentrations did correlate with shorter survival in acute lymphoblastic leukemia (p = 0.03), and with shorter complete remission duration in acute myelogenous leukemia (p = 0.04) and MDS (p = 0.03). CONCLUSION Circulating CD33 can be detected in the plasma from patients with leukemias, and cCD33 levels may have clinical implication, e.g., predictive and prognostic value, in these patients.
Collapse
Affiliation(s)
- Adam Abdool
- Hematology and Oncology Department, Quest Diagnostics Nichols Institute, San Juan Capistrano, CA 92675, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
O’Brien MM, Lacayo NJ, Lum BL, Kshirsagar S, Buck S, Ravindranath Y, Bernstein M, Weinstein H, Chang MN, Arceci RJ, Sikic BI, Dahl GV. Phase I study of valspodar (PSC-833) with mitoxantrone and etoposide in refractory and relapsed pediatric acute leukemia: a report from the Children's Oncology Group. Pediatr Blood Cancer 2010; 54:694-702. [PMID: 20209646 PMCID: PMC2838930 DOI: 10.1002/pbc.22366] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND Valspodar, a non-immunosuppressive analog of cylosporine, is a potent P-glycoprotein (MDR1) inhibitor. As MDR1-mediated efflux of chemotherapeutic agents from leukemic blasts may contribute to drug resistance, a phase 1 study of valspodar combined with mitoxantrone and etoposide in pediatric patients with relapsed or refractory leukemias was performed. PROCEDURE Patients received a valspodar-loading dose (2 mg/kg) followed by a 5-day continuous valspodar infusion (8, 10, 12.5, or 15 mg/kg/day) combined with lower than standard doses of mitoxantrone and etoposide. The valspodar dose was escalated using a standard 3 + 3 phase I design. RESULTS Twenty-one patients were evaluable for toxicity and 20 for response. The maximum tolerated dose (MTD) of valspodar was 12.5 mg/kg/day, combined with 50% dose-reduced mitoxantrone and etoposide. The clearance of mitoxantrone and etoposide was decreased by 64% and 60%, respectively, when combined with valspodar. Dose-limiting toxicities included stomatitis, ataxia, and bone marrow aplasia. Three of 11 patients with acute lymphoblastic leukemia (ALL) had complete responses while no patient with acute myeloid leukemia (AML) had an objective response. In vitro studies demonstrated P-glycoprotein expression on the blasts of 5 of 14 patients, although only 1 had inhibition of rhodamine efflux by valspodar. CONCLUSIONS While this regimen was tolerable, responses in this heavily pretreated population were limited to a subset of patients with ALL.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors
- ATP Binding Cassette Transporter, Subfamily B, Member 1/blood
- Adolescent
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Child
- Child, Preschool
- Cyclosporins/administration & dosage
- Cyclosporins/adverse effects
- Cyclosporins/pharmacokinetics
- Cyclosporins/therapeutic use
- Drug Resistance, Multiple
- Drug-Related Side Effects and Adverse Reactions
- Etoposide/administration & dosage
- Female
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Male
- Maximum Tolerated Dose
- Mitoxantrone/administration & dosage
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Recurrence
- Salvage Therapy
- Young Adult
Collapse
Affiliation(s)
- Maureen M. O’Brien
- Department of Pediatrics, Division of Hematology/Oncology, Stanford University School of Medicine, Stanford, CA
| | - Norman J. Lacayo
- Department of Pediatrics, Division of Hematology/Oncology, Stanford University School of Medicine, Stanford, CA
| | - Bert L. Lum
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA
- Department of Medicine, Division of Clinical Pharmacology, Stanford University School of Medicine, Stanford, CA
| | - Smita Kshirsagar
- Department of Medicine, Division of Clinical Pharmacology, Stanford University School of Medicine, Stanford, CA
| | - Steven Buck
- Children’s Hospital of Michigan, Detroit, MI
| | | | | | | | | | - Robert J. Arceci
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Branimir I. Sikic
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA
- Department of Medicine, Division of Clinical Pharmacology, Stanford University School of Medicine, Stanford, CA
| | - Gary V. Dahl
- Department of Pediatrics, Division of Hematology/Oncology, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
49
|
Petrich T, Korkmaz Z, Krull D, Frömke C, Meyer GJ, Knapp WH. In vitro experimental 211At-anti-CD33 antibody therapy of leukaemia cells overcomes cellular resistance seen in vivo against gemtuzumab ozogamicin. Eur J Nucl Med Mol Imaging 2010; 37:851-61. [DOI: 10.1007/s00259-009-1356-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Accepted: 12/01/2009] [Indexed: 11/28/2022]
|
50
|
Abstract
Over the past decade, monoclonal antibodies have dramatically impacted the treatment of haematological malignancies, as evidenced by the effect of rituximab on the response rate and survival of patients with follicular and diffuse large B cell non-Hodgkin's lymphoma. Currently, only two monoclonal antibodies - the anti-CD33 immunotoxin gemtuzumab ozogamicin and the CD52-directed antibody alemtuzumab - are approved for treatment of relapsed acute myeloid leukaemia in older patients and B cell chronic lymphocytic leukaemia, respectively. Although not approved for such treatment, alemtuzumab is also active against T cell prolymphocytic leukaemia, cutaneous T cell lymphoma and Sézary syndrome, and adult T cell leukaemia and lymphoma. In addition, rituximab has demonstrated activity against B cell chronic lymphocytic and hairy cell leukaemia. Monoclonal antibodies targeting CD4, CD19, CD20, CD22, CD23, CD25, CD45, CD66 and CD122 are now being studied in the clinic for the treatment of leukaemia. Here, we discuss how these new antibodies have been engineered to reduce immunogenicity and improve antibody targeting and binding. Improved interactions with Fc receptors on immune effector cells can enhance destruction of target cells through antibody-dependent cellular cytotoxicity and complement-mediated cell lysis. The antibodies can also be armed with cellular toxins or radionuclides to enhance the destruction of leukaemia cells.
Collapse
Affiliation(s)
- John C Morris
- Metabolism Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892-1457, USA.
| | | |
Collapse
|