Antibody-Drug Conjugates of Calicheamicin Derivative: Gemtuzumab Ozogamicin and Inotuzumab Ozogamicin

Total Page:16

File Type:pdf, Size:1020Kb

Antibody-Drug Conjugates of Calicheamicin Derivative: Gemtuzumab Ozogamicin and Inotuzumab Ozogamicin CCR FOCUS Antibody-Drug Conjugates of Calicheamicin Derivative: Gemtuzumab Ozogamicin and Inotuzumab Ozogamicin Alejandro D. Ricart Abstract Antibody-drug conjugates (ADC) are an attractive approach for the treatment of acute myeloid leukemia and non-Hodgkin lymphomas, which in most cases, are inherently sensitive to cytotoxic agents. CD33 and CD22 are specific markers of myeloid leukemias and B-cell malignancies, respectively. These endocytic receptors are ideal for an ADC strategy because they can effectively carry the cytotoxic payload into the cell. Gemtuzumab ozogamicin (GO, Mylotarg) and inotuzumab ozogamicin consist of a derivative of calichea- micin (a potent DNA-binding cytotoxic antibiotic) linked to a humanized monoclonal IgG4 antibody directed against CD33 or CD22, respectively. Both of these ADCs have a target-mediated pharmacokinetic disposition. GO was the first drug to prove the ADC concept in the clinic, specifically in phase II studies that included substantial proportions of older patients with relapsed acute myeloid leukemia. In contrast, in phase III studies, it has thus far failed to show clinical benefit in first-line treatment in combination with standard chemotherapy. Inotuzumab ozogamicin has shown remarkable clinical activity in relapsed/refractory B-cell non-Hodgkin lymphoma, and it has started phase III evaluation. The safety profile of these ADCs includes reversible myelosuppression (especially neutropenia and thrombocyto- penia), elevated hepatic transaminases, and hyperbilirubinemia. There have been postmarketing reports of hepatotoxicity, especially veno-occlusive disease, associated with GO. The incidence is 2%, but patients who undergo hematopoietic stem cell transplantation have an increased risk. As we steadily move toward the goal of personalized medicine, these kinds of agents will provide a unique opportunity to treat selected patient subpopulations based on the expression of their specific tumor targets. Clin Cancer Res; 17(20); 6417–27. Ó2011 AACR. Introduction The sialic-acid–binding immunoglobulin-like lectins (siglecs) include a family of receptors that are restrictedly It is surprising that few monoclonal antibodies (mAb) expressed on one or a few immune cell types, making them have been approved for the treatment of acute myeloid attractive for targeted therapy (8, 9). CD33 (also known as leukemia (AML) and non-Hodgkin lymphoma (NHL) since siglec-3) and CD22 (siglec-2) were identified as markers of the first approval of rituximab in 1997. These malignancies myeloid leukemias and B-cell lymphomas, respectively, localize in areas that are readily accessible to circulating almost 25 years ago. Siglecs are endocytic receptors and mAbs, such as the circulating blood, lymph nodes, and thus ideal for an ADC strategy because they can effectively bone marrow. The disposition of intact mAbs is also favor- carry the complex with its cytotoxic payload into the cell (8). able for their treatment: Distribution into other tissues is This is a striking difference from other antibody targets, such slow, and volumes of distribution are generally low (1). as CD20, for which human cells are highly heterogeneous in Antibody-drug conjugates (ADC) are an attractive approach their internalization ability (10). for the treatment of these diseases, which in most cases are inherently sensitive to cytotoxic agents. This CCR Focus Gemtuzumab Ozogamicin section describes a significant number of ADCs currently in clinical development (2–7). Description Gemtuzumab ozogamicin (GO, Mylotarg; Fig. 1) consists of a semisynthetic derivative of calicheamicin (N-acetyl-g calicheamicin 1,2-dimethyl hydrazine dichloride), a potent Author's Affiliation: Biotechnology Unit and Oncology Clinical Research, Pfizer Inc., San Diego, California enediyne DNA-binding cytotoxic antibiotic, linked to an engineered humanized monoclonal IgG4 antibody Corresponding Author: Alejandro D. Ricart, Biotechnology Unit and Oncology Clinical Research, La Jolla Laboratories, Pfizer Inc., 10646 (hP67.6) directed against the CD33 antigen present on Science Center Drive (CB1), San Diego, CA 92121. Phone: 858-622- leukemic myeloblasts in most patients with AML 7575; Fax: 877-481-3011; E-mail: alejandro.d.ricart@pfizer.com (80%). IgG4 has interesting properties for a carrier. It has doi: 10.1158/1078-0432.CCR-11-0486 the longest circulating half-life of all isotypes, with limited Ó2011 American Association for Cancer Research. ability for complement fixation and antibody-dependent www.aacrjournals.org 6417 Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research. CCR FOCUS A CH3 Figure 1. A, immunoconjugate binding and internalization. B, immunoconjugate intracellular trafficking from endosome to lysosome, with linker cleavage. Acid-labile AcBut hydrazone linker is cleaved in the acid environment of lysosome. C, calicheamicin derivative is released intracellularly. The reduction to the active enediyne form requires glutathione. D, the active enediyne form binds to the minor groove in DNA and causes double-strand breaks, resulting in cell death. E, Pgp- mediated efflux may be a mechanism of drug resistance in leukemic cells. The immunoconjugate structure on the upper right is modified from Advani et al. (52). Reprinted with permission. Ó 2008 American Society of Clinical Oncology. D cellular toxicity. The unconjugated antibody hP67.6 is not cells in vitro. This effective intracellular hydrolytic release of known to be cytotoxic per se (11). Calicheamicin was the calicheamicin derivative was important for its intracel- isolated from the actinomycete Micromonospora echinospora lular trafficking and the subsequent access to its DNA target. calichensis (12). The cytotoxic drug is attached to the anti- This indicates the importance of designing linkers that are body through a covalent linkage (condensation) of a specific for the individual target cell type (11). bifunctional linker, 4-(4-acetylphenoxy)butanoic acid (AcBut linker), which allows stability in physiologic Pharmacodynamics buffers (pH 7.4) and efficient calicheamicin release inside Saturation and internalization were analyzed in a phase II lysosomes (pH 4; ref. 11). The average loading of cali- study (13). Within 3 to 6 hours after a 9 mg/m2 infusion of cheamicin on the antibody is 2.5 mol/mol (drug-loading GO, nearly complete saturation of CD33 antigens was range of 2–3 mol of calicheamicin per mole of antibody; reached for leukemic and normal myeloid cells in blood. ref. 11). Calicheamicin binds to the minor groove in the Although the mean maximal binding of GO to myeloid DNA and causes double-strand DNA breaks, resulting in cell blast cells was comparable to monocytes, the maximal death (please refer to Fig. 1 to follow this sequence; ref. 12). binding to granulocytes was significantly lower, with no The custom-made, well-controlled, hydrolysable bond with binding to lymphocytes. These data are in accordance with the AcBut linker showed significantly more potent and the known expression levels of the CD33 antigen on these selective calicheamicin conjugates of P67.6 against HL-60 cells. On the other hand, this target is not (considerably) 6418 Clin Cancer Res; 17(20) October 15, 2011 Clinical Cancer Research Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research. Antibody-Drug Conjugates of Calicheamicin Derivative þ expressed on normal CD34 pluripotent hematopoietic (RP2D) was determined to be 9 mg/m2 infused i.v. over 2 stem cells or on nonhematopoietic tissues (see "Safety" hours, repeated on day 14 (16). Increased serum concentra- section for gemtuzumab ozogamicin below; ref. 14). Satu- tions were observed after the second dose and were believed ration levels before the second infusion were increased to be due to a decrease in clearance by CD33-positive blast compared with baseline levels (before the first infusion) in cells, a result of the lower peripheral leukemic burden after that phase II study, apparently due to remaining GO from the first dose (21, 22). The concentration profiles of cali- the first infusion. Upon binding to CD33, GO was rapidly cheamicin derivative followed the same time course as internalized, as determined by the decrease in maximal hP67.6, evidence that the payload remained conjugated to membrane binding (13). In vitro studies using pulse label- the antibody and was delivered to the leukemic cells. No ing with GO showed a continuous renewal of CD33, which relationship was found between plasma concentration and could increase the internalization and thereby the intracel- response at the RP2D (21). Despite high interpatient vari- lular accumulation of the cytotoxic agent (13). Using a ability, the pharmacokinetic parameters of the components lentivirus-mediated gene transfer to manipulate CD33 (hP67.6, total and unconjugated calicheamicin derivative) expression in cell lines that normally lack CD33 or have were not different based on gender or age, including a very low levels of CD33, Walter and colleagues (15) showed comparison between pediatric patients and adults (22, a quantitative relationship between CD33 expression and 23). Weight and body surface area did not affect the phar- GO-induced cytotoxicity. The CD33 cytoplasmic immunor- macokinetics of GO (23). Although most mAbs in oncology eceptor tyrosine-based inhibitory motifs controlled the are administered on the basis of body weight or surface area, internalization process, and point mutations that disrupted
Recommended publications
  • C-1027, a Radiomimetic Enediyne Anticancer Drug, Preferentially Targets Hypoxic Cells
    Research Article C-1027, A Radiomimetic Enediyne Anticancer Drug, Preferentially Targets Hypoxic Cells Terry A. Beerman,1 Loretta S. Gawron,1 Seulkih Shin,1 Ben Shen,2 and Mary M. McHugh1 1Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York;and 2Division of Pharmaceutical Sciences, University of Wisconsin National Cooperative Drug Discovery Group, and Department of Chemistry, University of Wisconsin, Madison, Wisconsin Abstract identified primarily from studies with neocarzinostatin (NCS), a The hypoxic nature of cells within solid tumors limits the holo-form drug, consisting of an apoprotein carrier and an active efficacy of anticancer therapies such as ionizing radiation and chromophore, and was assumed to be representative of all agents conventional radiomimetics because their mechanisms re- in this class (11). The NCS chromophore contains a bicyclic quire oxygen to induce lethal DNA breaks. For example, the enediyne that damages DNA via a Myers-Saito cycloaromatization conventional radiomimetic enediyne neocarzinostatin is 4- reaction, resulting in a 2,6-indacene diradical structure capable of fold less cytotoxic to cells maintained in low oxygen (hypoxic) hydrogen abstractions from deoxyribose (12, 13). Subsequent to compared with normoxic conditions. By contrast, the ene- generation of a sugar radical, reaction with oxygen quickly and diyne C-1027 was nearly 3-fold more cytotoxic to hypoxic than efficiently leads to formation of hydroxyl radicals that induce to normoxic cells. Like other radiomimetics, C-1027 induced DSBs/SSBs at a 1:5 ratio. The more recently discovered holo-form DNA breaks to a lesser extent in cell-free, or cellular hypoxic, enediyne C-1027 (Fig.
    [Show full text]
  • Pharmacologic Considerations in the Disposition of Antibodies and Antibody-Drug Conjugates in Preclinical Models and in Patients
    antibodies Review Pharmacologic Considerations in the Disposition of Antibodies and Antibody-Drug Conjugates in Preclinical Models and in Patients Andrew T. Lucas 1,2,3,*, Ryan Robinson 3, Allison N. Schorzman 2, Joseph A. Piscitelli 1, Juan F. Razo 1 and William C. Zamboni 1,2,3 1 University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA; [email protected] (J.A.P.); [email protected] (J.F.R.); [email protected] (W.C.Z.) 2 Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; [email protected] 3 Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; [email protected] * Correspondence: [email protected]; Tel.: +1-919-966-5242; Fax: +1-919-966-5863 Received: 30 November 2018; Accepted: 22 December 2018; Published: 1 January 2019 Abstract: The rapid advancement in the development of therapeutic proteins, including monoclonal antibodies (mAbs) and antibody-drug conjugates (ADCs), has created a novel mechanism to selectively deliver highly potent cytotoxic agents in the treatment of cancer. These agents provide numerous benefits compared to traditional small molecule drugs, though their clinical use still requires optimization. The pharmacology of mAbs/ADCs is complex and because ADCs are comprised of multiple components, individual agent characteristics and patient variables can affect their disposition. To further improve the clinical use and rational development of these agents, it is imperative to comprehend the complex mechanisms employed by antibody-based agents in traversing numerous biological barriers and how agent/patient factors affect tumor delivery, toxicities, efficacy, and ultimately, biodistribution.
    [Show full text]
  • Enediynes, Enyneallenes, Their Reactions, and Beyond
    Advanced Review Enediynes, enyne-allenes, their reactions, and beyond Elfi Kraka∗ and Dieter Cremer Enediynes undergo a Bergman cyclization reaction to form the labile 1,4-didehy- drobenzene (p-benzyne) biradical. The energetics of this reaction and the related Schreiner–Pascal reaction as well as that of the Myers–Saito and Schmittel reac- tions of enyne-allenes are discussed on the basis of a variety of quantum chemical and available experimental results. The computational investigation of enediynes has been beneficial for both experimentalists and theoreticians because it has led to new synthetic challenges and new computational methodologies. The accurate description of biradicals has been one of the results of this mutual fertilization. Other results have been the computer-assisted drug design of new antitumor antibiotics based on the biological activity of natural enediynes, the investigation of hetero- and metallo-enediynes, the use of enediynes in chemical synthesis and C materials science, or an understanding of catalyzed enediyne reactions. " 2013 John Wiley & Sons, Ltd. How to cite this article: WIREs Comput Mol Sci 2013. doi: 10.1002/wcms.1174 INTRODUCTION symmetry-allowed pericyclic reactions, (ii) aromatic- ity as a driving force for chemical reactions, and (iii) review on the enediynes is necessarily an ac- the investigation of labile intermediates with biradical count of intense and successful interdisciplinary A character. The henceforth called Bergman cyclization interactions of very different fields in chemistry provided deeper insight into the electronic structure involving among others organic chemistry, matrix of biradical intermediates, the mechanism of organic isolation spectroscopy, quantum chemistry, biochem- reactions, and orbital symmetry rules.
    [Show full text]
  • Inotuzumab Ozogamicin Is an Investigational Agent and Has Not Been Approved for Marketing by Any Regulatory Agency at This Time
    Inotuzumab Ozogamicin is an investigational agent and has not been approved for marketing by any regulatory agency at this time. ABOUT INOTUZUMAB OZOGAMICIN Inotuzumab ozogamicin is an investigational antibody-drug conjugate (ADC) comprised of a monoclonal antibody (mAb) targeting CD22, a cell surface antigen found on cancer cells in almost all B-ALL patients, linked to a cytotoxic agent.1,2 When inotuzumab ozogamicin binds to the CD22 antigen on B-cells, it is internalized into the cell, where the cytotoxic agent calicheamicin is released to destroy the cell.3 ABOUT ACUTE LYMPHOBLASTIC LEUKEMIA (ALL) Acute lymphoblastic leukemia (ALL) in adults is an aggressive leukemia that can be fatal within a matter of months if left untreated.4 While many potential treatments have been studied, only a limited number of medicines for relapsed or refractory adults with ALL have been approved by the FDA and other regulatory authorities in the past decade. The current standard treatment is intensive, lengthy chemotherapy with the goal of halting the signs and symptoms of ALL (called hematologic remission) and patients becoming eligible for a stem cell transplant.4 UNMET NEEDS IN ADULT ALL Statistics & Patient Characteristics Statistics & Patient Characteristics • In 2017, it is estimated that 5,970 cases of ALL • Approximately half of U.S. adults who learn they will be diagnosed in the United States, with about have ALL this year will not respond to commonly 4 in 10 cases occurring in adults.5 used chemotherapy agents (e.g., be refractory) or will eventually see their disease return (e.g., relapse).5 • While significant strides have been made in treating children with ALL, the opposite is true for adults with • The outlook for adults with relapsed or refractory the disease.
    [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]
  • Whither Radioimmunotherapy: to Be Or Not to Be? Damian J
    Published OnlineFirst April 20, 2017; DOI: 10.1158/0008-5472.CAN-16-2523 Cancer Perspective Research Whither Radioimmunotherapy: To Be or Not To Be? Damian J. Green1,2 and Oliver W. Press1,2,3 Abstract Therapy of cancer with radiolabeled monoclonal antibodies employing multistep "pretargeting" methods, particularly those has produced impressive results in preclinical experiments and in utilizing bispecific antibodies, have greatly enhanced the thera- clinical trials conducted in radiosensitive malignancies, particu- peutic efficacy of radioimmunotherapy and diminished its toxi- larly B-cell lymphomas. Two "first-generation," directly radiola- cities. The dramatically improved therapeutic index of bispecific beled anti-CD20 antibodies, 131iodine-tositumomab and 90yttri- antibody pretargeting appears to be sufficiently compelling to um-ibritumomab tiuxetan, were FDA-approved more than a justify human clinical trials and reinvigorate enthusiasm for decade ago but have been little utilized because of a variety of radioimmunotherapy in the treatment of malignancies, particu- medical, financial, and logistic obstacles. Newer technologies larly lymphomas. Cancer Res; 77(9); 1–6. Ó2017 AACR. "To be, or not to be, that is the question: Whether 'tis nobler in the pembrolizumab (anti-PD-1), which are not directly cytotoxic mind to suffer the slings and arrows of outrageous fortune, or to take for cancer cells but "release the brakes" on the immune system, arms against a sea of troubles, And by opposing end them." Hamlet. allowing cytotoxic T cells to be more effective at recognizing –William Shakespeare. and killing cancer cells. Outstanding results have already been demonstrated with checkpoint inhibiting antibodies even in far Introduction advanced refractory solid tumors including melanoma, lung cancer, Hodgkin lymphoma and are under study for a multi- Impact of monoclonal antibodies on the field of clinical tude of other malignancies (4–6).
    [Show full text]
  • Monoclonal Antibodies
    MONOCLONAL ANTIBODIES ALEMTUZUMAB ® (CAMPATH 1H ) I. MECHANISM OF ACTION Antibody-dependent lysis of leukemic cells following cell surface binding. Alemtuzumab is a recombinant DNA-derived humanized monoclonal antibody that is directed against surface glycoprotein CD52. CD52 is expressed on the surface of normal and malignant B and T lymphocytes, NK cells, monocytes, macrophages, a subpopulation of granulocytes, and tissues of the male reproductive system (CD 52 is not expressed on erythrocytes or hematopoietic stem cells). The alemtuzumab antibody is an IgG1 kappa with human variable framework and constant regions, and complementarity-determining regions from a murine monoclonal antibody (campath 1G). II. PHARMACOKINETICS Cmax and AUC show dose proportionality over increasing dose ranges. The overall average half-life is 12 days. Peak and trough levels of Campath rise during the first weeks of Campath therapy, and approach steady state by week 6. The rise in serum Campath concentration corresponds with the reduction in malignant lymphocytes. III. DOSAGE AND ADMINISTRATION Campath can be administered intravenously or subcutaneously. Intravenous: Alemtuzumab therapy should be initiated at a dose of 3 mg administered as a 2-hour IV infusion daily. When the 3 mg dose is tolerated (i.e., ≤ Grade 2 infusion related side effects), the daily dose should be escalated to 10mg and continued until tolerated (i.e., ≤ Grade 2 infusion related side effects). When the 10 mg dose is tolerated, the maintenance dose of 30 mg may be initiated. The maintenance dose of alemtuzumab is 30 mg/day administered three times a week on alternate days (i.e. Monday, Wednesday, and Friday), for up to 12 weeks.
    [Show full text]
  • 1826 ALL FINAL 2[2].Pptx
    7/17/18 Treatment Challenges in Acute Lymphoblastic Leukemia Pharmacists’ Key Roles in Ensuring Best Practices This educational activity is sponsored by Postgraduate Healthcare Education, LLC and supported by an educational grant from Pfizer. 1 7/17/18 Faculty Christopher A. Fausel, PharmD, MHA, BCOP Clinical Manager, Oncology Pharmacy Indiana University Health Chairman, Hoosier Cancer Research Network Indianapolis, Indiana Dr. Fausel is the Clinical Manager of the Oncology Pharmacy at the Indiana University, Simon Cancer Center in Indianapolis and oversees clinical and dispensing activities for oncology pharmacy services at Indiana University Health. He also serves as Chairman of the Board of the Hoosier Cancer Research Network, a non-profit organization that conducts clinical trials and translational research in cancer and is the administrative center of the Big Ten Cancer Research Consortium. Dr. Fausel is the Chair of a biomedical IRB and serves on the IRB Executive Committee for Indiana University. He is a long standing member of ASHP, ASCO and HOPA. He has authored numerous academic writings and is a nationally invited speaker on topics of oncology therapeutics and oncology pharmacy practice. Disclosures Dr. Fausel has no relevant affiliations or financial relationships with a commercial interest to disclose. The clinical reviewer, Megan May, PharmD, BCOP, has no actual or potential conflict of interest in relation to this program. Susanne Batesko, RN, BSN, Robin Soboti, RPh, and Susan R. Grady, MSN, RN-BC, as well as the planners, managers, and other individuals, not previously disclosed, who are in a position to control the content of Postgraduate Healthcare Education (PHE) continuing education activities hereby state that they have no relevant conflicts of interest and no financial relationships or relationships to products or devices during the past 12 months to disclose in relation to this activity.
    [Show full text]
  • Inotuzumab Ozogamicin) for Injection, for Intravenous Use for Injection: 0.9 Mg Lyophilized Powder in a Single-Dose Vial for Initial U.S
    HIGHLIGHTS OF PRESCRIBING INFORMATION • See full prescribing information for instructions on reconstitution of These highlights do not include all the information needed to use lyophilized powder, and preparation and administration of reconstituted BESPONSA™ safely and effectively. See full prescribing information for drug. (2.4) BESPONSA. --------------------- DOSAGE FORMS AND STRENGTHS---------------------­ BESPONSA (inotuzumab ozogamicin) for injection, for intravenous use For injection: 0.9 mg lyophilized powder in a single-dose vial for Initial U.S. Approval: 2017 reconstitution and further dilution. (3) WARNING: HEPATOTOXICITY, INCLUDING HEPATIC VENO­ ------------------------------ CONTRAINDICATIONS -----------------------------­ OCCLUSIVE DISEASE (VOD) (ALSO KNOWN AS SINUSOIDAL None (4) OBSTRUCTION SYNDROME and INCREASED RISK OF POST­ HEMATOPOIETIC STEM CELL TRANSPLANT (HSCT) NON­ ----------------------- WARNINGS AND PRECAUTIONS ----------------------­ RELAPSE MORTALITY • Myelosuppression: Monitor complete blood counts; for signs and See full prescribing information for complete boxed warning. symptoms of infection; bleeding/hemorrhage; or other effects of myelosuppression during treatment; manage appropriately. (5.3) • Hepatotoxicity, including fatal and life-threatening VOD occurred in • Infusion related reactions: Monitor for infusion related reactions during and patients who received BESPONSA. (5.1) for at least 1 hour after infusion ends. (5.4) • A higher post-HSCT non-relapse mortality rate occurred in patients • QT interval
    [Show full text]
  • F. Gherlinzoni CD33: Gemtuzumab Ozogamicin
    1ST CUNEO CITY IMMUNOTHERAPY CONFERENCE 17-18 Maggio 2018 PASSIVE IMMUNOTHERAPY ANTIBODY-DRUG CONJUGATES GEMTUZUMAB OZOGAMICIN Do#. Filippo Gherlinzoni Direore Unità Operava Complessa di Ematologia Ospedale “Ca’ Foncello” - Treviso IL RIECCOLO Blood Reviews 28 (2014) 143–153 Contents lists available at ScienceDirect Blood Reviews journal homepage: www.elsevier.com/locate/blre REVIEW The past and future of CD33 as therapeutic target in acute myeloid leukemia a a,b a,b,c, George144 S. Laszlo , Elihu H. Estey , Roland B. Walter ⁎G.S. Laszlo et al. / Blood Reviews 28 (2014) 143–153 a Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA b Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA c Department of Epidemiology, University of Washington, Seattle, WA, USA as cytokine-induced cellular proliferation [14,33]. Engagement with CD33 antibodies has similarly been shown to affect cytokine and article info abstract chemokine secretion by monocytes, although both increases [34] or Keywords: CD33 is a myeloid differentiation antigen with endocytic properties.decreases It is broadly[33] expressedhave on been acute myeloidobserved in vitro. Acute myeloid leukemia leukemia (AML) blasts and, possibly, some leukemic stem cells and has therefore been exploited as target for Antibody therapeutic antibodies for many years. The improved survival seen in many patients when the antibody-drug Antibody-drug conjugate conjugate, gemtuzumab ozogamicin, is added to conventional chemotherapy validates this approach. However, fi 2.3. Endocytic properties of CD33 Bispeci c antibody many attempts with unconjugated or conjugated antibodies have been unsuccessful, highlighting the challenges BiTE of targeting CD33 in AML.
    [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]
  • Immunotherapies Shape the Treatment Landscape for Hematologic Malignancies Jane De Lartigue, Phd
    Feature Immunotherapies shape the treatment landscape for hematologic malignancies Jane de Lartigue, PhD he treatment landscape for hematologic of TIL therapy has been predominantly limited to malignancies is evolving faster than ever melanoma.1,3,4 before, with a range of available therapeutic Most recently, there has been a substantial buzz Toptions that is now almost as diverse as this group around the idea of genetically engineering T cells of tumors. Immunotherapy in particular is front and before they are reintroduced into the patient, to center in the battle to control these diseases. Here, increase their anti-tumor efficacy and minimize we describe the latest promising developments. damage to healthy tissue. This is achieved either by manipulating the antigen binding portion of the Exploiting T cells T-cell receptor to alter its specificity (TCR T cells) The treatment landscape for hematologic malig- or by generating artificial fusion receptors known as nancies is diverse, but one particular type of therapy chimeric antigen receptors (CAR T cells; Figure 1). has led the charge in improving patient outcomes. The former is limited by the need for the TCR to be Several features of hematologic malignancies may genetically matched to the patient’s immune type, make them particularly amenable to immunother- whereas the latter is more flexible in this regard and apy, including the fact that they are derived from has proved most successful. corrupt immune cells and come into constant con- CARs are formed by fusing part of the single- tact with other immune cells within the hemato- chain variable fragment of a monoclonal antibody poietic environment in which they reside.
    [Show full text]