Antibody–Drug Conjugates for Cancer Therapy

Total Page:16

File Type:pdf, Size:1020Kb

Antibody–Drug Conjugates for Cancer Therapy molecules Review Antibody–Drug Conjugates for Cancer Therapy Umbreen Hafeez 1,2,3, Sagun Parakh 1,2,3 , Hui K. Gan 1,2,3,4 and Andrew M. Scott 1,3,4,5,* 1 Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC 3084, Australia; [email protected] (U.H.); [email protected] (S.P.); [email protected] (H.K.G.) 2 Department of Medical Oncology, Olivia Newton-John Cancer and Wellness Centre, Austin Health, Melbourne, VIC 3084, Australia 3 School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia 4 Department of Medicine, University of Melbourne, Melbourne, VIC 3084, Australia 5 Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC 3084, Australia * Correspondence: [email protected]; Tel.: +61-39496-5000 Academic Editor: João Paulo C. Tomé Received: 14 August 2020; Accepted: 13 October 2020; Published: 16 October 2020 Abstract: Antibody–drug conjugates (ADCs) are novel drugs that exploit the specificity of a monoclonal antibody (mAb) to reach target antigens expressed on cancer cells for the delivery of a potent cytotoxic payload. ADCs provide a unique opportunity to deliver drugs to tumor cells while minimizing toxicity to normal tissue, achieving wider therapeutic windows and enhanced pharmacokinetic/pharmacodynamic properties. To date, nine ADCs have been approved by the FDA and more than 80 ADCs are under clinical development worldwide. In this paper, we provide an overview of the biology and chemistry of each component of ADC design. We briefly discuss the clinical experience with approved ADCs and the various pathways involved in ADC resistance. We conclude with perspectives about the future development of the next generations of ADCs, including the role of molecular imaging in drug development. Keywords: antibody–drug conjugate; ADC; monoclonal antibody; cytotoxic payload; linkers; cancer; molecular imaging 1. Introduction Despite the range of new anti-cancer drugs in the market, there were 9.6 million deaths from cancer globally in 2018, with approximately one in six of all deaths being due to cancer [1]. A recent review concluded that most of the anti-cancer drugs that entered the market between 2009 and 2013 showed only marginal gains in terms of overall survival and that there is an urgent need to increase therapeutic efficacy [2]. Systemic therapies based on the use of monoclonal antibodies (mAbs) started to emerge after the discovery of hybridoma technology by Kohler and Milstein in 1975. In 1988, Greg Winter pioneered the technique to humanize monoclonal antibodies and thereafter, therapeutic monoclonal antibodies have been successfully developed for the treatment of various cancers. To date, approximately 30 mAbs have been approved by the US Food and Drug Administration (FDA) for the treatment of cancer. The specificity of mAbs also makes them well suited for the targeted delivery of drugs to cancer cells whilst avoiding normal tissues. Monoclonal antibody technology has been further improved over the next decades by conjugating antibodies with cytotoxic drugs [3]. Such conjugates, called antibody–drug conjugates (ADCs), are targeted agents that link a cytotoxic drug (also called as cytotoxic payload or warhead) via a linker to a monoclonal antibody which specifically recognizes a cellular surface antigen and deliver toxic payload at the tumor site, thus improving the efficacy of chemotherapy and reducing systemic exposure and toxicity [4]. More than 80 ADCs are currently under clinical development worldwide [5,6]. In this paper, we provide an overview of the biology and Molecules 2020, 25, 4764; doi:10.3390/molecules25204764 www.mdpi.com/journal/molecules Molecules 2020, 25, 4764 2 of 35 Molecules 2020, 25, 4764 2 of 33 biology and chemistry of ADCs, discuss key components of ADC design and discuss the current statuschemistry of clinically of ADCs, approved discuss key and components underdeveloped of ADC ADCs. design We and also discuss discuss the resistance current status mechanisms of clinically to ADC,approved the role and underdevelopedof molecular imaging ADCs. in Wethe alsoclinic discussal development resistance of mechanisms ADCs and give to ADC, an opinion the role on of futuremolecular perspectives. imaging in the clinical development of ADCs and give an opinion on future perspectives. 2. Design Design and Structure of Antibody–Drug Conjugates The useuse ofof ADCs ADCs has has evolved evolved over over time, time, becoming becoming increasingly increasingly widespread widespread as initial as initial problems problems were wereovercome overcome by better by targetbetter selection, target selection, accompanied acco bympanied improvements by improvements in payloads, in linker payloads, technologies linker technologiesand conjugation andtechniques conjugation (Figure techniques1). (Figure 1). Figure 1. Antibody–drug conjugate structure. Figure 1. Antibody–drug conjugate structure. 2.1. Target Selection 2.1. TargetUnconjugated Selection mAbs have various mechanisms of action including target receptor neutralization, receptorUnconjugated downregulation, mAbs signalling have disruption,various mechanisms antibody dependent of action cell-mediated including cytotoxicity target receptor (ADCC), neutralization,complement-dependent receptor downregu cell-mediatedlation, cytotoxicity signalling (CDCC) disruption, and immuneantibody checkpoint dependent inhibition cell-mediated [7,8]. cytotoxicityOn the other (ADCC), hand, ADCs complement-dependent rely on target receptor ce internalizationll-mediated cytotoxicity to deliver the(CDCC) cytotoxic and payload immune to checkpointthe cancer cells.inhibition Hence, [7,8]. selecting On the an other appropriate hand, ADCs target rely antigen on target is areceptor critical stepinternalization for the success to deliver of an theantibody–drug cytotoxic payload conjugate. to the cancer cells. Hence, selecting an appropriate target antigen is a critical step forIdeally, the success the target of an antigen antibody–drug should be conjugate. abundant on the cell surface to be available for binding by theIdeally, circulating the target ADC, antigen e.g., melanoma should be cellabundant lines withon the p97 cell receptor surface expressionto be available levels for from binding 80,000 by theto 280,000 circulating receptors ADC, per e.g., cell melanoma showed sensitivity cell lines with to ADC p97 L49-vcMMAF, receptor expression while otherlevels cancer from 80,000 cell lines to with280,000 lower receptors p97 expression per cell showed were resistantsensitivity to to L49-vcMMAF ADC L49-vcMMAF, [9]. However, while other various cancer other cell ADCs lines havewith lowershown p97 effi expressioncacy over a were wide resistant range of to antigen L49-vcMMAF expression [9]. However, levels based various on other other characteriztics ADCs have shown of the efficacytarget antigen, over a includingwide range binding of anti agenffinity expression and rate levels of internalization. based on other Gemtuzumab characteriztics ozogamicin of the target has antigen,demonstrated including efficacy binding at a relatively affinity lowand expressionrate of internalization. of CD33 (5000 Gemtuzumab to 10,000 receptors ozogamicin per cell) has as demonstrated efficacy at a relatively low expression of CD33 (5000 to 10,000 receptors per cell) as Molecules 2020, 25, 4764 3 of 33 compared to trastuzumab emtansine (T-DM1) which usually requires high ErbB2 expression levels (>2 million receptors per cell) [10,11]. The high surface expression of the antigen may not lead to ADC efficacy if trafficking into the tumor cell is impaired. Inge et al. [12] showed that after antibody binding CD21 does not effectively internalize, even when expressed at very high levels, resulting in a lack of efficacy with anti-CD21-MCC-DM1 ADCs. This study also showed that CD21 forms a complex with CD19 on the surface of B cells and prevents internalization of CD19 after it is bound to anti-CD19 antibody and decreasing anti-CD19-MCC-DM1 efficacy [12]. To minimize off-target toxicity, the target antigen should have exclusive or preferential expression on cancer cells with a minimal expression on healthy tissue. Four out of nine approved ADCs—inotuzumab ozogamicin, gemtuzumab ozogamicin, brentuximab vedotin, and polatuzumab vedotin—target lineage-specific markers that include CD22, CD33, CD30 and CD79, respectively. These antigens have consistent expression across the target cell population. The target antigen should also have minimal secretion in circulation from the cancer cells, as antibodies can bind to these secreted receptors in the circulation, thus limiting the amount of antibody available for target cell binding [13]. The target antigen should internalize efficiently upon ADC binding to allow the antigen–ADC complex to be internalized via receptor-mediated endocytosis, and it should undergo appropriate intracellular trafficking and degradation, thereby allowing the cytotoxic warhead to be released [14]. Target antigens on the cell surface can have variable rates for basal and antibody induced internalization which can affect ADC efficacy [12]. For example, CD74 antigen is rapidly internalized into target cells following antibody binding. The preclinical data for anti-CD74 ADC immu-110 showed similar efficacy to other ADCs that target slower internalizing antigens with more potent cytotoxic payloads [15]. Inadequate or inefficient internalization increases the chance of toxicity from inappropriate payload delivery outside the cancer cell
Recommended publications
  • Genmab and ADC Therapeutics Announce Amended Agreement for Camidanlumab Tesirine (Cami)
    Genmab and ADC Therapeutics Announce Amended Agreement for Camidanlumab Tesirine (Cami) Media Release Copenhagen, Denmark and Lausanne, Switzerland, October 30, 2020 • ADC Therapeutics to continue the development and commercialization of Cami • Genmab to receive mid-to-high single-digit tiered royalty Genmab A/S (Nasdaq: GMAB) and ADC Therapeutics SA (NYSE: ADCT) today announced that they have executed an amended agreement for ADC Therapeutics to continue the development and commercialization of camidanlumab tesirine (Cami). The parties first entered into a collaboration and license agreement in June 2013 for the development of Cami, an antibody drug conjugate (ADC) which combines Genmab’s HuMax®-TAC antibody targeting CD25 with ADC Therapeutics’ highly potent pyrrolobenzodiazepine (PBD) warhead technology. Under the terms of the 2013 agreement, the parties were to determine the path forward for continued development and commercialization of Cami upon completion of a Phase 1a/b clinical trial. ADC Therapeutics previously announced that Cami achieved an overall response rate of 86.5%, including a complete response rate of 48.6%, in Hodgkin lymphoma patients in this trial who had received a median of five prior lines of therapy. Cami is currently being evaluated in a 100-patient pivotal Phase 2 clinical trial intended to support the submission of a Biologics License Application (BLA) to the U.S. Food and Drug Administration (FDA). The trial is more than 50 percent enrolled and ADC Therapeutics anticipates reporting interim results in the first half of 2021. “We have a long-standing relationship with the ADC Therapeutics team and believe they are an ideal partner for the ongoing development and potential commercialization of Cami,” said Jan van de Winkel, Ph.D., Chief Executive Officer of Genmab.
    [Show full text]
  • Draft Minutes PDCO 12-15 November 2019
    11 December 2019 EMA/PDCO/615413/2019 Inspections, Human Medicines Pharmacovigilance and Committees Division Paediatric Committee (PDCO) Minutes for the meeting on 12-15 November 2019 Chair: Koenraad Norga – Vice-Chair: Sabine Scherer Health and safety information In accordance with the Agency’s health and safety policy, delegates are to be briefed on health, safety and emergency information and procedures prior to the start of the meeting. Disclaimers Some of the information contained in these minutes is considered commercially confidential or sensitive and therefore not disclosed. With regard to intended therapeutic indications or procedure scopes listed against products, it must be noted that these may not reflect the full wording proposed by applicants and may also vary during the course of the review. Additional details on some of these procedures will be published in the PDCO Committee meeting reports (after the PDCO Opinion is adopted), and on the Opinions and decisions on paediatric investigation plans webpage (after the EMA Decision is issued). Of note, this set of minutes is a working document primarily designed for PDCO members and the work the Committee undertakes. Further information with relevant explanatory notes can be found at the end of this document. Note on access to documents Some documents mentioned in these minutes cannot be released at present following a request for access to documents within the framework of Regulation (EC) No 1049/2001 as they are subject to on- going procedures for which a final decision has not yet been adopted. They will become public when adopted or considered public according to the principles stated in the Agency policy on access to documents (EMA/127362/2006).
    [Show full text]
  • Safety and Antitumor Activity Study Evaluating Loncastuximab Tesirine and Rituximab Versus Immunochemotherapy in Diffuse Large B-Cell Lymphoma
    Safety and Antitumor Activity Study Evaluating Loncastuximab Tesirine and Rituximab Versus Immunochemotherapy in Diffuse Large B-Cell Lymphoma Yuliya Linhares1, Mitul Gandhi2, Michael Chung3, Jennifer Adeleye4, David Ungar4, Mehdi Hamadani5 1Medical Oncology, Miami Cancer Institute, Baptist Health, Miami, FL, USA; 2Medical Oncology, Virginia Cancer Specialists, Gainesville, VA, USA; 3Hematology/Oncology, The Oncology Institute of Hope and Innovation, Downey, CA, USA; 4Clinical Development, ADC Therapeutics America, Inc., Murray Hill, NJ, USA; 5Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA Poster slides, 62nd ASH Annual Meeting and Poster session II, Sunday, December 6, 2020: Exposition, Virtual Meeting, December 5–8, 2020 7:00 am – 3:30 pm (Pacific Time) Introduction The prognosis of patients with DLBCL whose disease is refractory to initial chemotherapy (and are thus ineligible for ASCT) or relapse early after ASCT is extremely poor1,2 The development of a more effective, less toxic salvage treatment for DLBCL remains an unmet need2 Loncastuximab tesirine (Lonca) is an ADC comprising a Mechanism of action of Lonca humanized monoclonal anti-CD19 antibody conjugated to a pyrrolobenzodiazepine (PBD) dimer toxin, through a protease cleavable valine–alanine linker Rituximab is an anti-CD20 monoclonal antibody used as a standard component of care for the treatment of DLBCL, either as monotherapy or in combination with chemotherapy In a Phase 2 study, Lonca demonstrated single-agent antitumor activity with manageable toxicity in patients with R/R DLBCL3 Rituximab is licensed for treatment of NHL but is being used in combination with an unlicensed drug (loncastuximab tesirine) in this study 1. Crump M, et al.
    [Show full text]
  • Antibody–Drug Conjugates
    Published OnlineFirst April 12, 2019; DOI: 10.1158/1078-0432.CCR-19-0272 Review Clinical Cancer Research Antibody–Drug Conjugates: Future Directions in Clinical and Translational Strategies to Improve the Therapeutic Index Steven Coats1, Marna Williams1, Benjamin Kebble1, Rakesh Dixit1, Leo Tseng1, Nai-Shun Yao1, David A. Tice1, and Jean-Charles Soria1,2 Abstract Since the first approval of gemtuzumab ozogamicin nism of activity of the cytotoxic warhead. However, the (Mylotarg; Pfizer; CD33 targeted), two additional antibody– enthusiasm to develop ADCs has not been dampened; drug conjugates (ADC), brentuximab vedotin (Adcetris; Seat- approximately 80 ADCs are in clinical development in tle Genetics, Inc.; CD30 targeted) and inotuzumab ozogami- nearly 600 clinical trials, and 2 to 3 novel ADCs are likely cin (Besponsa; Pfizer; CD22 targeted), have been approved for to be approved within the next few years. While the hematologic cancers and 1 ADC, trastuzumab emtansine promise of a more targeted chemotherapy with less tox- (Kadcyla; Genentech; HER2 targeted), has been approved to icity has not yet been realized with ADCs, improvements treat breast cancer. Despite a clear clinical benefit being dem- in technology combined with a wealth of clinical data are onstrated for all 4 approved ADCs, the toxicity profiles are helping to shape the future development of ADCs. In this comparable with those of standard-of-care chemotherapeu- review, we discuss the clinical and translational strategies tics, with dose-limiting toxicities associated with the mecha- associated with improving the therapeutic index for ADCs. Introduction in antibody, linker, and warhead technologies in significant depth (2, 3, 8, 9). Antibody–drug conjugates (ADC) were initially designed to leverage the exquisite specificity of antibodies to deliver targeted potent chemotherapeutic agents with the intention of improving Overview of ADCs in Clinical Development the therapeutic index (the ratio between the toxic dose and the Four ADCs have been approved over the last 20 years (Fig.
    [Show full text]
  • Antibody–Drug Conjugates: the Last Decade
    pharmaceuticals Review Antibody–Drug Conjugates: The Last Decade Nicolas Joubert 1,* , Alain Beck 2 , Charles Dumontet 3,4 and Caroline Denevault-Sabourin 1 1 GICC EA7501, Equipe IMT, Université de Tours, UFR des Sciences Pharmaceutiques, 31 Avenue Monge, 37200 Tours, France; [email protected] 2 Institut de Recherche Pierre Fabre, Centre d’Immunologie Pierre Fabre, 5 Avenue Napoléon III, 74160 Saint Julien en Genevois, France; [email protected] 3 Cancer Research Center of Lyon (CRCL), INSERM, 1052/CNRS 5286/UCBL, 69000 Lyon, France; [email protected] 4 Hospices Civils de Lyon, 69000 Lyon, France * Correspondence: [email protected] Received: 17 August 2020; Accepted: 10 September 2020; Published: 14 September 2020 Abstract: An armed antibody (antibody–drug conjugate or ADC) is a vectorized chemotherapy, which results from the grafting of a cytotoxic agent onto a monoclonal antibody via a judiciously constructed spacer arm. ADCs have made considerable progress in 10 years. While in 2009 only gemtuzumab ozogamicin (Mylotarg®) was used clinically, in 2020, 9 Food and Drug Administration (FDA)-approved ADCs are available, and more than 80 others are in active clinical studies. This review will focus on FDA-approved and late-stage ADCs, their limitations including their toxicity and associated resistance mechanisms, as well as new emerging strategies to address these issues and attempt to widen their therapeutic window. Finally, we will discuss their combination with conventional chemotherapy or checkpoint inhibitors, and their design for applications beyond oncology, to make ADCs the magic bullet that Paul Ehrlich dreamed of. Keywords: antibody–drug conjugate; ADC; bioconjugation; linker; payload; cancer; resistance; combination therapies 1.
    [Show full text]
  • 5.01.582 Antibody-Drug Conjugates
    MEDICAL POLICY – 5.01.582 Antibody-Drug Conjugates Effective Date: June 1, 2021 RELATED MEDICAL POLICIES: Last Revised: May 11, 2021 None Replaces: N/A Select a hyperlink below to be directed to that section. POLICY CRITERIA | CODING | RELATED INFORMATION EVIDENCE REVIEW | REFERENCES | HISTORY ∞ Clicking this icon returns you to the hyperlinks menu above. Introduction An antibody is a blood protein. When the immune system detects an unhealthy cell, antibodies link to a molecule, known as an antigen, on the unhealthy cell. Monoclonal antibodies are produced in a laboratory. They are made to link to antigens usually found in high numbers on cancer cells. Antibody-drug conjugates combine monoclonal antibodies with certain chemotherapy drugs. The monoclonal antibodies find the cancer cells and the chemotherapy drug is released directly into those cells. The goal with this treatment is to target only cancer cells and spare nearby healthy cells. This policy describes when specific antibody-drug conjugates may be considered medically necessary. Note: The Introduction section is for your general knowledge and is not to be taken as policy coverage criteria. The rest of the policy uses specific words and concepts familiar to medical professionals. It is intended for providers. A provider can be a person, such as a doctor, nurse, psychologist, or dentist. A provider also can be a place where medical care is given, like a hospital, clinic, or lab. This policy informs them about when a service may be covered. Policy Coverage Criteria Drug Medical
    [Show full text]
  • Antibodies for the Treatment of Brain Metastases, a Dream Or a Reality?
    pharmaceutics Review Antibodies for the Treatment of Brain Metastases, a Dream or a Reality? Marco Cavaco, Diana Gaspar, Miguel ARB Castanho * and Vera Neves * Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal * Correspondence: [email protected] (M.A.R.B.C.); [email protected] (V.N.) Received: 19 November 2019; Accepted: 28 December 2019; Published: 13 January 2020 Abstract: The incidence of brain metastases (BM) in cancer patients is increasing. After diagnosis, overall survival (OS) is poor, elicited by the lack of an effective treatment. Monoclonal antibody (mAb)-based therapy has achieved remarkable success in treating both hematologic and non-central-nervous system (CNS) tumors due to their inherent targeting specificity. However, the use of mAbs in the treatment of CNS tumors is restricted by the blood–brain barrier (BBB) that hinders the delivery of either small-molecules drugs (sMDs) or therapeutic proteins (TPs). To overcome this limitation, active research is focused on the development of strategies to deliver TPs and increase their concentration in the brain. Yet, their molecular weight and hydrophilic nature turn this task into a challenge. The use of BBB peptide shuttles is an elegant strategy. They explore either receptor-mediated transcytosis (RMT) or adsorptive-mediated transcytosis (AMT) to cross the BBB. The latter is preferable since it avoids enzymatic degradation, receptor saturation, and competition with natural receptor substrates, which reduces adverse events. Therefore, the combination of mAbs properties (e.g., selectivity and long half-life) with BBB peptide shuttles (e.g., BBB translocation and delivery into the brain) turns the therapeutic conjugate in a valid approach to safely overcome the BBB and efficiently eliminate metastatic brain cells.
    [Show full text]
  • Loncastuximab Tesirine-Lpyl
    HIGHLIGHTS OF PRESCRIBING INFORMATION These highlights do not include all the information needed to use ZYNLONTA safely and effectively. See full prescribing information for ZYNLONTA. ZYNLONTA™ (loncastuximab tesirine-lpyl) for injection, for ------------------------WARNINGS AND PRECAUTIONS----------------------- intravenous use Effusion and Edema: Monitor for the development of pleural effusion, Initial U.S. Approval: 2021 pericardial effusion, ascites, peripheral edema, and general edema. Consider diagnostic imaging when symptoms develop or worsen. (5.1) -----------------------------INDICATIONS AND USAGE-------------------------- Myelosuppression: Monitor blood cell counts. Withhold, reduce, or ZYNLONTA is a CD19-directed antibody and alkylating agent conjugate discontinue ZYNLONTA based on severity. (5.2) indicated for the treatment of adult patients with relapsed or refractory large Infections: Monitor for infection and treat promptly. (5.3) B-cell lymphoma after two or more lines of systemic therapy, including Cutaneous Reactions: Monitor patients for new or worsening cutaneous diffuse large B-cell lymphoma (DLBCL) not otherwise specified, DLBCL reactions, including photosensitivity reactions. Dermatologic consultation arising from low grade lymphoma, and high-grade B-cell lymphoma. (1) should be considered. (5.4) Embryo-Fetal Toxicity: Can cause fetal harm. Advise patients of the This indication is approved under accelerated approval based on overall potential risk to a fetus and to use effective contraception. (5.5, 8.1, 8.3)
    [Show full text]
  • Loncastuximab Tesirine), a Novel Pyrrolobenzodiazepine-Based Antibody–Drug Conjugate, in Relapsed/ Refractory B-Cell Non-Hodgkin Lymphoma Brad S
    Published OnlineFirst November 4, 2019; DOI: 10.1158/1078-0432.CCR-19-0711 Clinical Trials: Targeted Therapy Clinical Cancer Research A Phase I Study of ADCT-402 (Loncastuximab Tesirine), a Novel Pyrrolobenzodiazepine-Based Antibody–Drug Conjugate, in Relapsed/ Refractory B-Cell Non-Hodgkin Lymphoma Brad S. Kahl1, Mehdi Hamadani2, John Radford3, Carmelo Carlo-Stella4, Paolo Caimi5, Erin Reid6, Jay M. Feingold7, Kirit M. Ardeshna8, Melhem Solh9, Leonard T. Heffner10, David Ungar7, Shui He7, Joseph Boni7, Karin Havenith11, and Owen A. O'Connor12 Abstract Purpose: ADCT-402 (loncastuximab tesirine) is an anti- 200 mg/kg. Treatment-emergent adverse events (TEAEs) were body–drug conjugate comprising a CD19-targeting antibody experienced by 87/88 (98.9%) patients. Most common TEAEs and pyrrolobenzodiazepine dimers. A first-in-human study (20% of patients) were hematologic abnormalities, fatigue, evaluated the safety and preliminary clinical activity of lon- edema, liver test abnormalities, nausea, rash, and dyspnea. castuximab tesirine in patients with B-cell non-Hodgkin lym- Grade 3 TEAEs (5% of patients) included hematologic phoma (NHL). abnormalities, liver test abnormalities, fatigue, and dyspnea. Experimental Design: A multicenter, phase I, dose- Overall response rate at doses 120 mg/kg was 59.4% (41 of 69 escalation and dose-expansion study enrolled patients ages patients; 40.6% complete response; 18.8% partial response). 18 years with relapsed/refractory (R/R) B-cell NHL. Patients Median duration of response, progression-free survival, and received loncastuximab tesirine every 3 weeks at doses overall survival (all doses) were 4.8, 5.5, and 11.6 months, assigned by a 3þ3 dose-escalation design.
    [Show full text]
  • Antibodies to Watch in 2021 Hélène Kaplona and Janice M
    MABS 2021, VOL. 13, NO. 1, e1860476 (34 pages) https://doi.org/10.1080/19420862.2020.1860476 PERSPECTIVE Antibodies to watch in 2021 Hélène Kaplona and Janice M. Reichert b aInstitut De Recherches Internationales Servier, Translational Medicine Department, Suresnes, France; bThe Antibody Society, Inc., Framingham, MA, USA ABSTRACT ARTICLE HISTORY In this 12th annual installment of the Antibodies to Watch article series, we discuss key events in antibody Received 1 December 2020 therapeutics development that occurred in 2020 and forecast events that might occur in 2021. The Accepted 1 December 2020 coronavirus disease 2019 (COVID-19) pandemic posed an array of challenges and opportunities to the KEYWORDS healthcare system in 2020, and it will continue to do so in 2021. Remarkably, by late November 2020, two Antibody therapeutics; anti-SARS-CoV antibody products, bamlanivimab and the casirivimab and imdevimab cocktail, were cancer; COVID-19; Food and authorized for emergency use by the US Food and Drug Administration (FDA) and the repurposed Drug Administration; antibodies levilimab and itolizumab had been registered for emergency use as treatments for COVID-19 European Medicines Agency; in Russia and India, respectively. Despite the pandemic, 10 antibody therapeutics had been granted the immune-mediated disorders; first approval in the US or EU in 2020, as of November, and 2 more (tanezumab and margetuximab) may Sars-CoV-2 be granted approvals in December 2020.* In addition, prolgolimab and olokizumab had been granted first approvals in Russia and cetuximab saratolacan sodium was first approved in Japan. The number of approvals in 2021 may set a record, as marketing applications for 16 investigational antibody therapeutics are already undergoing regulatory review by either the FDA or the European Medicines Agency.
    [Show full text]
  • Treating Non-Hodgkin Lymphoma If You’Ve Been Diagnosed with Non-Hodgkin Lymphoma, Your Treatment Team Will Discuss Your Options with You
    cancer.org | 1.800.227.2345 Treating Non-Hodgkin Lymphoma If you’ve been diagnosed with non-Hodgkin lymphoma, your treatment team will discuss your options with you. It’s important to weigh the benefits of each treatment option against the possible risks and side effects. How is non-Hodgkin lymphoma treated? Depending on the type and stage (extent) of the lymphoma and other factors, treatment options for people with NHL might include: ● Chemotherapy for Non-Hodgkin Lymphoma ● Immunotherapy for Non-Hodgkin Lymphoma ● Targeted Drug Therapy for Non-Hodgkin Lymphoma ● Radiation Therapy for Non-Hodgkin Lymphoma ● High-Dose Chemotherapy and Stem Cell Transplant for Non-Hodgkin Lymphoma ● Surgery for Non-Hodgkin Lymphoma Common treatment approaches Treatment approaches for NHL depend on the type of cancer, how advanced it is, as well as your health and other factors. Another important part of treatment for many people is palliative or supportive care. This can help prevent or treat problems such as infections, low blood cell counts, or some symptoms caused by the lymphoma. ● Treating B-Cell Non-Hodgkin Lymphoma ● Treating T-Cell Non-Hodgkin Lymphoma ● Treating HIV-Associated Lymphoma 1 ____________________________________________________________________________________American Cancer Society cancer.org | 1.800.227.2345 ● Palliative and Supportive Care for Non-Hodgkin Lymphoma Who treats non-Hodgkin lymphoma? Based on your treatment options, you may have different types of doctors on your treatment team. These doctors could include: ● A medical oncologist or hematologist: a doctor who treats lymphoma with chemotherapy, immunotherapy, and targeted therapy. ● A radiation oncologist: a doctor who treats cancer with radiation therapy. ● A bone marrow transplant doctor: a doctor who specializes in treating cancer or other diseases with bone marrow or stem cell transplants.
    [Show full text]
  • Antibody–Drug Conjugates for Cancer Therapy
    molecules Review Antibody–Drug Conjugates for Cancer Therapy Umbreen Hafeez 1,2,3, Sagun Parakh 1,2,3 , Hui K. Gan 1,2,3,4 and Andrew M. Scott 1,3,4,5,* 1 Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC 3084, Australia; [email protected] (U.H.); [email protected] (S.P.); [email protected] (H.K.G.) 2 Department of Medical Oncology, Olivia Newton-John Cancer and Wellness Centre, Austin Health, Melbourne, VIC 3084, Australia 3 School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia 4 Department of Medicine, University of Melbourne, Melbourne, VIC 3084, Australia 5 Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC 3084, Australia * Correspondence: [email protected]; Tel.: +61-39496-5000 Academic Editor: João Paulo C. Tomé Received: 14 August 2020; Accepted: 13 October 2020; Published: 16 October 2020 Abstract: Antibody–drug conjugates (ADCs) are novel drugs that exploit the specificity of a monoclonal antibody (mAb) to reach target antigens expressed on cancer cells for the delivery of a potent cytotoxic payload. ADCs provide a unique opportunity to deliver drugs to tumor cells while minimizing toxicity to normal tissue, achieving wider therapeutic windows and enhanced pharmacokinetic/pharmacodynamic properties. To date, nine ADCs have been approved by the FDA and more than 80 ADCs are under clinical development worldwide. In this paper, we provide an overview of the biology and chemistry of each component of ADC design. We briefly discuss the clinical experience with approved ADCs and the various pathways involved in ADC resistance.
    [Show full text]