1
|
Badar T, Luger SM, Litzow MR. Incorporation of immunotherapy into frontline treatment for adults with B-cell precursor acute lymphoblastic leukemia. Blood 2025; 145:1475-1484. [PMID: 39236292 DOI: 10.1182/blood.2023022921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 09/07/2024] Open
Abstract
ABSTRACT Although complete remission rates in adults with B-cell precursor acute lymphoblastic leukemia (BCP-ALL) have improved over the last 2 decades, it is still inferior to that of the pediatric population, and once in remission, the risk of relapse is still high. Furthermore, although pediatric-inspired chemotherapy regimens have improved long-term outcomes for adolescents and young adults, these intensive chemotherapy regimens are not well tolerated in older patients and are associated with higher morbidity and mortality. Immunotherapeutic agents offer a potential opportunity to improve response and decrease relapse without increasing toxicity. The incorporation of rituximab (anti-CD20 monoclonal antibody) into chemotherapy regimens has been shown to improve outcomes. The treatment of BCP-ALL in adults has been transformed with the approval of inotuzumab ozogamicin (anti-CD22 antibody-drug conjugate), blinatumomab (CD3/CD19 bispecific antibody construct), and chimeric antigen receptor T cells for relapsed or refractory disease and of blinatumomab for measurable residual disease (MRD)-positive remission. More recently, studies of inotuzumab and blinatumomab have shown promising results when used up front either with or without multiagent chemotherapy. Blinatumomab has also been shown in a randomized trial to provide a survival benefit in patients with MRD-negative first remission when added to chemotherapy, which recently led to its additional US Food and Drug Administration approval for use in consolidation. In this review, we highlight the evolution of chemoimmunotherapy-based treatment approaches in the management of treatment-naïve BCP-ALL.
Collapse
Affiliation(s)
- Talha Badar
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapy Program, Mayo Clinic, Jacksonville, FL
| | - Selina M Luger
- Abramson Cancer Center and Division of Hematology-Oncology, University of Pennsylvania, Philadelphia, PA
| | | |
Collapse
|
2
|
Abstract
Background: The introduction of monoclonal antibodies, either as native molecules or conjugated to radioisotopes or other toxins, has led to new therapeutic options for patients with hematologic malignancies. In addition, the use of small molecules against specific cell surface receptors, enzymes, and proteins has become an important strategy in the treatment of such disorders. Methods: The author reviewed the published clinical trials of monoclonal antibody and other targeted therapies in hematologic malignancies. Results: Results from several trials demonstrate a therapeutic benefit for the use of monoclonal antibodies (either native or conjugated) and other targeted therapies, used alone or in combination with standard cytotoxic chemotherapy. Conclusions: Targeted therapy of hematologic malignancies seems to be an effective and less toxic approach to the treatment of such disorders. Nevertheless, additional studies are needed to determine where and when such management fits into a therapeutic regimen for any given disorder, whether upfront or as salvage therapy, alone or in combination with chemotherapy (concurrent or sequential).
Collapse
MESH Headings
- Antibodies, Monoclonal/therapeutic use
- Antineoplastic Agents/classification
- Antineoplastic Agents/therapeutic use
- Drug Delivery Systems/trends
- Hematologic Neoplasms/drug therapy
- Hematologic Neoplasms/immunology
- Hematologic Neoplasms/therapy
- Humans
- Immunologic Factors/immunology
- Immunologic Factors/therapeutic use
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
- Lymphoma, Non-Hodgkin/drug therapy
- Lymphoma, Non-Hodgkin/immunology
- Multiple Myeloma/drug therapy
- Multiple Myeloma/immunology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology
- Radioimmunotherapy
Collapse
Affiliation(s)
- Philip Kuriakose
- Department of Internal Medicine, Division of Hematology/Oncology, Henry Ford Hospital, Detroit, MI 48202, USA.
| |
Collapse
|
3
|
Zhou Y, Li XP, Chen BY, Tumer NE. Ricin uses arginine 235 as an anchor residue to bind to P-proteins of the ribosomal stalk. Sci Rep 2017; 7:42912. [PMID: 28230053 PMCID: PMC5322317 DOI: 10.1038/srep42912] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 01/17/2017] [Indexed: 01/24/2023] Open
Abstract
Ricin toxin A chain (RTA) binds to stalk P-proteins to reach the α-sarcin/ricin loop (SRL) where it cleaves a conserved adenine. Arginine residues at the RTA/RTB interface are involved in this interaction. To investigate the individual contribution of each arginine, we generated single, double and triple arginine mutations in RTA. The R235A mutation reduced toxicity and depurination activity more than any other single arginine mutation in yeast. Further reduction in toxicity, depurination activity and ribosome binding was observed when R235A was combined with a mutation in a nearby arginine. RTA interacts with the ribosome via a two-step process, which involves slow and fast interactions. Single arginine mutations eliminated the fast interactions with the ribosome, indicating that they increase the binding rate of RTA. Arginine residues form a positively charged patch to bind to negatively charged residues at the C-termini of P-proteins. When electrostatic interactions conferred by the arginines are lost, hydrophobic interactions are also abolished, suggesting that the hydrophobic interactions alone are insufficient to allow binding. We propose that Arg235 serves as an anchor residue and cooperates with nearby arginines and the hydrophobic interactions to provide the binding specificity and strength in ribosome targeting of RTA.
Collapse
Affiliation(s)
- Yijun Zhou
- Department of Plant Biology and Pathology, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, New Jersey 08901-8520, USA
| | - Xiao-Ping Li
- Department of Plant Biology and Pathology, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, New Jersey 08901-8520, USA
| | - Brian Y Chen
- Department of Computer Science and Engineering, Lehigh University, Bethlehem, Pennsylvania 18015-3084, USA
| | - Nilgun E Tumer
- Department of Plant Biology and Pathology, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, New Jersey 08901-8520, USA
| |
Collapse
|
4
|
Maino E, Bonifacio M, Scattolin AM, Bassan R. Immunotherapy approaches to treat adult acute lymphoblastic leukemia. Expert Rev Hematol 2016; 9:563-77. [DOI: 10.1586/17474086.2016.1170593] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
5
|
Krishnan V, Xu X, Kelly D, Snook A, Waldman SA, Mason RW, Jia X, Rajasekaran AK. CD19-Targeted Nanodelivery of Doxorubicin Enhances Therapeutic Efficacy in B-Cell Acute Lymphoblastic Leukemia. Mol Pharm 2015; 12:2101-11. [PMID: 25898125 DOI: 10.1021/acs.molpharmaceut.5b00071] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Nanomedicine has advanced to clinical trials for adult cancer therapy. However, the field is still in its infancy for treatment of childhood malignancies such as acute lymphoblastic leukemia (ALL). Nanotherapy offers multiple advantages over conventional therapy. It facilitates targeted delivery and enables controlled release of drugs to reduce treatment-related side effects. Here, we demonstrate that doxorubicin (DOX) encapsulated in polymeric nanoparticles (NPs) modified with targeting ligands against CD19 (CD19-DOX-NPs) can be delivered in a CD19-specific manner to leukemic cells. The CD19-DOX-NPs were internalized via receptor-mediated endocytosis and imparted cytotoxicity in a CD19-dependent manner in CD19-positive ALL cells. Leukemic mice treated with CD19-DOX-NPs survived significantly longer and manifested a higher degree of agility, indicating reduced apparent systemic toxicity during treatment compared to mice treated with free DOX. We suggest that targeted delivery of drugs used in childhood cancer treatment should improve therapeutic efficacy and reduce treatment-related side effects in children.
Collapse
Affiliation(s)
- Vinu Krishnan
- ⊥Nemours Center for Childhood Cancer Research, A. I. duPont Hospital for Children, Wilmington, Delaware 19803, United States
| | | | | | - Adam Snook
- #Pharmacology and Experimental Therapeutics, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, United States
| | - Scott A Waldman
- #Pharmacology and Experimental Therapeutics, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, United States
| | - Robert W Mason
- ⊥Nemours Center for Childhood Cancer Research, A. I. duPont Hospital for Children, Wilmington, Delaware 19803, United States
| | | | - Ayyappan K Rajasekaran
- ∇Therapy Architects, LLC, 2700 Silverside Road, Wilmington, Delaware 19810, United States
| |
Collapse
|
6
|
Maino E, Scattolin AM, Viero P, Sancetta R, Pascarella A, Vespignani M, Bassan R. Modern immunotherapy of adult B-lineage acute lymphoblastic leukemia with monoclonal antibodies and chimeric antigen receptor modified T cells. Mediterr J Hematol Infect Dis 2015; 7:e2015001. [PMID: 25574360 PMCID: PMC4283921 DOI: 10.4084/mjhid.2015.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 11/20/2014] [Indexed: 11/08/2022] Open
Abstract
The introduction of newer cytotoxic monoclonal antibodies and chimeric antigen receptor modified T cells is opening a new age in the management of B-lineage adult acute lymphoblastic leukemia. This therapeutic change must be very positively acknowledged because of the limits of intensive chemotherapy programs and allogeneic stem cell transplantation. In fact, with these traditional therapeutic tools the cure can be achieved in only 40-50% of the patients. The failure rates are particularly high in the elderly, in patients with post-induction persistence of minimal residual disease and especially in refractory/relapsed disease. The place of the novel immunotherapeutics in improving the outcome of adult patients with B-lineage acute lymphoblastic leukemia is reviewed.
Collapse
Affiliation(s)
- Elena Maino
- Hematology and Bone Marrow Transplant Unit, Ospedale dell’Angelo e SS. Giovanni e Paolo, Mestre-Venezia, Italy
| | - Anna Maria Scattolin
- Hematology and Bone Marrow Transplant Unit, Ospedale dell’Angelo e SS. Giovanni e Paolo, Mestre-Venezia, Italy
| | - Piera Viero
- Hematology and Bone Marrow Transplant Unit, Ospedale dell’Angelo e SS. Giovanni e Paolo, Mestre-Venezia, Italy
| | - Rosaria Sancetta
- Hematology and Bone Marrow Transplant Unit, Ospedale dell’Angelo e SS. Giovanni e Paolo, Mestre-Venezia, Italy
| | - Anna Pascarella
- Hematology and Bone Marrow Transplant Unit, Ospedale dell’Angelo e SS. Giovanni e Paolo, Mestre-Venezia, Italy
| | - Michele Vespignani
- Hematology and Bone Marrow Transplant Unit, Ospedale dell’Angelo e SS. Giovanni e Paolo, Mestre-Venezia, Italy
| | - Renato Bassan
- Hematology and Bone Marrow Transplant Unit, Ospedale dell’Angelo e SS. Giovanni e Paolo, Mestre-Venezia, Italy
| |
Collapse
|
7
|
Walker A, Mrózek K, Kohlschmidt J, Rao KW, Pettenati MJ, Sterling LJ, Marcucci G, Carroll AJ, Bloomfield CD. New recurrent balanced translocations in acute myeloid leukemia and myelodysplastic syndromes: cancer and leukemia group B 8461. Genes Chromosomes Cancer 2013; 52:385-401. [PMID: 23225546 PMCID: PMC3874732 DOI: 10.1002/gcc.22036] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 10/31/2012] [Indexed: 12/16/2022] Open
Abstract
Acquired chromosome abnormalities in patients with acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) are among the most valuable determinants of diagnosis and prognosis. In search of new recurrent balanced translocations, we reviewed the Cancer and Leukemia Group B (CALGB) cytogenetics database containing pretreatment and relapse karyotypes of 4,701 adults with AML and 565 with MDS who were treated on CALGB trials. We identified all cases with balanced structural rearrangements occurring as a sole abnormality or in addition to one other abnormality, excluded abnormalities known to be recurrent, and then reviewed the literature to determine whether any of what we considered unique, previously unknown abnormalities had been reported. As a result, we identified seven new recurrent balanced translocations in AML or MDS: t(7;11)(q22;p15.5), t(10;11)(q23;p15), t(2;12)(p13;p13), t(12;17)(p13;q12), t(2;3)(p21;p21), t(5;21)(q31;q22), and t(8;14)(q24.1;q32.2), and additionally, t(10;12)(p11;q15), a new translocation in AML previously reported in a case of acute lymphoblastic leukemia. Herein, we report hematologic and clinical characteristics and treatment outcomes of patients with these newly recognized recurrent translocations. We also report 52 unique balanced translocations, together with the clinical data of patients harboring them, which to our knowledge have not been previously published. We hope that once the awareness of their existence is increased, some of these translocations may become recognized as novel recurring abnormalities. Identification of additional cases with both the new recurrent and the unique balanced translocations will enable determination of their prognostic significance and help to provide insights into the mechanisms of disease pathogenesis in patients with these rare abnormalities.
Collapse
Affiliation(s)
- Alison Walker
- Division of Hematology and Oncology, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH
| | - Krzysztof Mrózek
- Division of Hematology and Oncology, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH
| | - Jessica Kohlschmidt
- Division of Hematology and Oncology, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN
| | - Kathleen W. Rao
- University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Mark J. Pettenati
- Comprehensive Cancer Center Wake Forest University, Winston-Salem, NC
| | - Lisa J. Sterling
- Division of Hematology and Oncology, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH
| | - Guido Marcucci
- Division of Hematology and Oncology, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH
| | | | - Clara D. Bloomfield
- Division of Hematology and Oncology, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH
| |
Collapse
|
8
|
Carol H, Szymanska B, Evans K, Boehm I, Houghton PJ, Smith MA, Lock RB. The anti-CD19 antibody-drug conjugate SAR3419 prevents hematolymphoid relapse postinduction therapy in preclinical models of pediatric acute lymphoblastic leukemia. Clin Cancer Res 2013; 19:1795-805. [PMID: 23426279 DOI: 10.1158/1078-0432.ccr-12-3613] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE Relapsed or refractory pediatric acute lymphoblastic leukemia (ALL) remains a major cause of death from cancer in children. In this study, we evaluated the efficacy of SAR3419, an antibody-drug conjugate of the maytansinoid DM4 and a humanized anti-CD19 antibody, against B-cell precursor (BCP)-ALL and infant mixed lineage leukemia (MLL) xenografts. EXPERIMENTAL DESIGN ALL xenografts were established as systemic disease in immunodeficient (NOD/SCID) mice from direct patient explants. SAR3419 was administered as a single agent and in combination with an induction-type regimen of vincristine/dexamethasone/l-asparaginase (VXL). Leukemia progression and response to treatment were assessed in real-time, and responses were evaluated using strict criteria modeled after the clinical setting. RESULTS SAR3419 significantly delayed the progression of 4 of 4 CD19(+) BCP-ALL and 3 of 3 MLL-ALL xenografts, induced objective responses in all but one xenograft but was ineffective against T-lineage ALL xenografts. Relative surface CD19 expression across the xenograft panel significantly correlated with leukemia progression delay and objective response measure scores. SAR3419 also exerted significant efficacy against chemoresistant BCP-ALL xenografts over a large (10-fold) dose range and significantly enhanced VXL-induced leukemia progression delay in two highly chemoresistant xenografts by up to 82 days. When administered as protracted therapy following remission induction with VXL, SAR3419 prevented disease recurrence into hematolymphoid and other major organs with the notable exception of central nervous system involvement. CONCLUSION These results suggest that incorporation of SAR3419 into remission induction protocols may improve the outcome for high-risk pediatric and adult CD19(+) ALL.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/pharmacology
- Antigens, CD19/genetics
- Antigens, CD19/metabolism
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/pharmacology
- Dose-Response Relationship, Drug
- Drug Evaluation, Preclinical
- Humans
- Induction Chemotherapy
- Maytansine/administration & dosage
- Maytansine/analogs & derivatives
- Maytansine/pharmacology
- Mice
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Recurrence
- Treatment Outcome
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Hernan Carol
- Children's Cancer Institute Australia for Medical Research, University of New South Wales, Sydney, Australia
| | | | | | | | | | | | | |
Collapse
|
9
|
Improving the Therapeutic Potential of Human Granzyme B for Targeted Cancer Therapy. Antibodies (Basel) 2013. [DOI: 10.3390/antib2010019] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|
10
|
|
11
|
Abstract
ALL blast cells express a variety of specific antigens e.g. CD19, CD20, CD22, CD33, and CD52, which serve as targets for Monoclonal Antibodies (MoAbs). So far, the most experience is available for anti-CD20 (rituximab), which has been combined with chemotherapy for treatment of mature B-ALL/Burkitt's lymphoma. Studies with rituximab have also been completed in B-precursor ALL. Another antigen, CD19, is of great interest due to a very high rate of expression in ALL. It can be targeted by a bispecific monoclonal antibody, Blinatumomab, directed against CD19 and CD3. Smaller studies or case reports are also available for the anti CD52 antibody (Alemtuzumab), for anti CD22 (Epratuzumab) or anti CD33 (Gemtuzumab). Available data demonstrate that MoAb therapy in ALL is a highly promising targeted treatment. However, several details for an optimal treatment approach e.g. the required level of antigen expression, timing, schedule, dosage and stage of disease still need to be defined.
Collapse
|
12
|
Dosio F, Brusa P, Cattel L. Immunotoxins and anticancer drug conjugate assemblies: the role of the linkage between components. Toxins (Basel) 2011; 3:848-83. [PMID: 22069744 PMCID: PMC3202854 DOI: 10.3390/toxins3070848] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 07/02/2011] [Accepted: 07/06/2011] [Indexed: 12/02/2022] Open
Abstract
Immunotoxins and antibody-drug conjugates are protein-based drugs combining a target-specific binding domain with a cytotoxic domain. Such compounds are potentially therapeutic against diseases including cancer, and several clinical trials have shown encouraging results. Although the targeted elimination of malignant cells is an elegant concept, there are numerous practical challenges that limit conjugates’ therapeutic use, including inefficient cellular uptake, low cytotoxicity, and off-target effects. During the preparation of immunoconjugates by chemical synthesis, the choice of the hinge component joining the two building blocks is of paramount importance: the conjugate must remain stable in vivo but must afford efficient release of the toxic moiety when the target is reached. Vast efforts have been made, and the present article reviews strategies employed in developing immunoconjugates, focusing on the evolution of chemical linkers.
Collapse
Affiliation(s)
- Franco Dosio
- Department of Drug Science and Technology, University of Torino, Torino 10125, Italy.
| | | | | |
Collapse
|
13
|
Ferreras JM, Citores L, Iglesias R, Jiménez P, Girbés T. Use of ribosome-inactivating proteins from Sambucus for the construction of immunotoxins and conjugates for cancer therapy. Toxins (Basel) 2011; 3:420-41. [PMID: 22069717 PMCID: PMC3202832 DOI: 10.3390/toxins3050420] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 04/02/2011] [Accepted: 04/25/2011] [Indexed: 01/28/2023] Open
Abstract
The type 2 ribosome-inactivating proteins (RIPs) isolated from some species belonging to the Sambucus genus, have the characteristic that although being even more active than ricin inhibiting protein synthesis in cell-free extracts, they lack the high toxicity of ricin and related type 2 RIPs to intact cells and animals. This is due to the fact that after internalization, they follow a different intracellular pathway that does not allow them to reach the cytosolic ribosomes. The lack of toxicity of type 2 RIPs from Sambucus make them good candidates as toxic moieties in the construction of immunotoxins and conjugates directed against specific targets. Up to now they have been conjugated with either transferrin or anti-CD105 to target either transferrin receptor- or endoglin-overexpressing cells, respectively.
Collapse
Affiliation(s)
- José M. Ferreras
- Department of Biochemistry, Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, E-47005 Valladolid, Spain; (L.C.); (R.I.)
| | - Lucía Citores
- Department of Biochemistry, Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, E-47005 Valladolid, Spain; (L.C.); (R.I.)
| | - Rosario Iglesias
- Department of Biochemistry, Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, E-47005 Valladolid, Spain; (L.C.); (R.I.)
| | - Pilar Jiménez
- Nutrition and Bromatology, Faculty of Medicine, E-47005 Valladolid, Spain; (P.J.); (T.G.)
| | - Tomás Girbés
- Nutrition and Bromatology, Faculty of Medicine, E-47005 Valladolid, Spain; (P.J.); (T.G.)
| |
Collapse
|
14
|
Hoelzer D. Novel antibody-based therapies for acute lymphoblastic leukemia. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2011; 2011:243-249. [PMID: 22160041 DOI: 10.1182/asheducation-2011.1.243] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
A major breakthrough in the treatment of acute lymphoblastic leukemia (ALL) was the availability of targeted therapies targeting either specific transcripts, such as bcr-abl fusion protein by tyrosine kinase inhibitors (TKIs), or specific antigens by mAbs. ALL blast cells express a variety of specific antigens (eg, CD19, CD20, CD22, CD33, and CD52) that serve as targets for mAbs. To date, the most data are available for anti-CD20 (rituximab), which has been combined with chemotherapy for the treatment of mature B-ALL/Burkitt lymphoma. Studies with rituximab have also been completed in B-precursor ALL. Another antigen, CD19, is of great interest due to a very high rate of expression in ALL. It can be targeted by a bispecific mAb, blinatumomab, directed against CD19 and CD3. Smaller studies or case reports are also available for the anti-CD52 (alemtuzumab), anti-CD22 (epratuzumab), and anti-CD33 (gemtuzumab) mAbs. Available data demonstrate that mAb therapy in ALL is a highly promising treatment approach. However, several details for an optimal treatment approach, such as the required level of antigen expression, timing, schedule, dosage, and stage of disease, still need to be defined.
Collapse
|
15
|
|
16
|
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
|
17
|
Bernt KM, Prokop A, Huebener N, Gaedicke G, Wrasidlo W, Lode HN. Eradication of CD19+ leukemia by targeted calicheamicin θ. Bioconjug Chem 2009; 20:1587-94. [PMID: 19572629 DOI: 10.1021/bc900128h] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Children with relapsed and refractory acute lymphoblastic leukemia (ALL) still face a critical prognosis. We tested the hypothesis that targeted calicheamicin theta (θ) using an anti-CD19-immunoconjugate may provide an effective treatment strategy for CD19(+) ALL. Calicheamicin θ is a rationally designed prodrug of the natural enediyene calicheamicin γ, obtained by total synthesis. It offers the advantage of increased in vivo stability and 1000-fold higher antitumor potency over calicheamicin γ. First, we demonstrate efficacy of calicheamicin θ against primary pre-B leukemic cells and multidrug-resistant leukemia cell lines (IC(50) = 10(-9) to 10(-12) M). Second, conjugation of calicheamicin θ to an internalizing murine anti-CD19 monoclonal antibody was demonstrated to affect neither calicheamicin θ mediated cytotoxicity nor binding of the antibody to the target molecule. Third, anti-CD19-calicheamicin θ immunoconjugate revealed a maximum tolerated dose of 10 μg/kg and CD19-specific and long-lasting eradication of established leukemia was demonstrated in a xenograft model. Finally, we show that the antileukemic effect of anti-CD19-calicheamicin θ is mediated by induction of apoptosis proceeding through the caspase-mediated mitochondrial pathway. On the basis of these results, we conclude that anti-CD19-calicheamicin θ immunoconjugates may offer a novel and effective approach for the treatment of relapsed CD19(+) ALL.
Collapse
Affiliation(s)
- Kathrin M Bernt
- Charité Universitätsmedizin Berlin, Department of Pediatrics, Experimental Oncology, Berlin, Germany
| | | | | | | | | | | |
Collapse
|
18
|
Kvinlaug BT, Lazic SE. Cancer stem cell patents. Expert Opin Ther Pat 2008. [DOI: 10.1517/13543770802553764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
19
|
Du X, Beers R, FitzGerald DJ, Pastan I. Differential cellular internalization of anti-CD19 and -CD22 immunotoxins results in different cytotoxic activity. Cancer Res 2008; 68:6300-5. [PMID: 18676854 PMCID: PMC2561922 DOI: 10.1158/0008-5472.can-08-0461] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
B-cell malignancies routinely express surface antigens CD19 and CD22. Immunotoxins against both antigens have been evaluated, and the immunotoxins targeting CD22 are more active. To understand this disparity in cytotoxicity and guide the screening of therapeutic targets, we compared two immunotoxins, FMC63(Fv)-PE38-targeting CD19 and RFB4(Fv)-PE38 (BL22)-targeting CD22. Six lymphoma cell lines have 4- to 9-fold more binding sites per cell for CD19 than for CD22, but BL22 is 4- to 140-fold more active than FMC63(Fv)-PE38, although they have a similar cell binding affinity (Kd, approximately 7 nmol/L). In 1 hour, large amounts of BL22 are internalized (2- to 3-fold more than the number of CD22 molecules on the cell surface), whereas only 5.2% to 16.6% of surface-bound FMC63(Fv)-PE38 is internalized. The intracellular reservoir of CD22 decreases greatly after immunotoxin internalization, indicating that it contributes to the uptake of BL22. Treatment of cells with cycloheximide does not reduce the internalization of BL22. Both internalized immunotoxins are located in the same vesicles. Our results show that the rapid internalization of large amounts of BL22 bound to CD22 makes CD22 a better therapeutic target than CD19 for immunotoxins and probably for other immunoconjugates that act inside cells.
Collapse
Affiliation(s)
- Xing Du
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Richard Beers
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | | |
Collapse
|
20
|
What determines the outcomes for adolescents and young adults with acute lymphoblastic leukemia treated on cooperative group protocols? A comparison of Children's Cancer Group and Cancer and Leukemia Group B studies. Blood 2008; 112:1646-54. [PMID: 18502832 DOI: 10.1182/blood-2008-01-130237] [Citation(s) in RCA: 414] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
We performed a retrospective comparison of presenting features, planned treatment, complete remission (CR) rate, and outcome of 321 adolescents and young adults (AYAs) ages 16 to 20 years with newly diagnosed acute lymphoblastic leukemia (ALL) who were treated on consecutive trials in either the Children's Cancer Group (CCG) or the Cancer and Leukemia Group B (CALGB) from 1988 to 2001. CR rates were identical, 90% for both CALGB and CCG AYAs. CCG AYAs had a 63% event-free survival (EFS) and 67% overall survival (OS) at 7 years in contrast to the CALGB AYAs, in which 7-year EFS was only 34% (P < .001; relative hazard rate [RHR] = 2.2) and OS was 46% (P < .001; RHR = 1.9). While CALGB AYAs aged 16 to 17 years achieved similar outcomes to all CCG AYAs with a 7-year EFS of 55%, the EFS for 18- to 20-year-old CALGB patients was only 29%. Comparison of the regimens showed that CCG AYAs received earlier and more intensive central nervous system prophylaxis and higher cumulative doses of nonmyelosuppressive agents. There were no differences in outcomes of those who reached maintenance therapy on time compared with those who were delayed. Based on these observations, a prospective study for AYAs with ALL using the more successful approach of the CCG has been initiated.
Collapse
|
21
|
Gojo I. Improving treatment strategies for acute lymphoblastic leukemia by combining immunotherapy and chemotherapy. Leuk Res 2008; 32:847-9. [PMID: 18191203 DOI: 10.1016/j.leukres.2007.11.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Revised: 11/28/2007] [Accepted: 11/29/2007] [Indexed: 11/25/2022]
|
22
|
|
23
|
Muñoz R, Arias Y, Ferreras JM, Rojo MA, Gayoso MJ, Nocito M, Benitez J, Jiménez P, Bernabéu C, Girbés T. Targeting a marker of the tumour neovasculature using a novel anti-human CD105-immunotoxin containing the non-toxic type 2 ribosome-inactivating protein nigrin b. Cancer Lett 2007; 256:73-80. [PMID: 17637501 DOI: 10.1016/j.canlet.2007.05.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Revised: 05/18/2007] [Accepted: 05/23/2007] [Indexed: 01/16/2023]
Abstract
Targeting tumour neovasculature using antibodies to the endothelial receptor CD105 (endoglin), is a potentially useful approach for anti-tumour therapy. We report on the preparation and the cytotoxicity of a novel immunotoxin consisting in the non-toxic type 2 ribosome-inactivating protein (RIP) nigrin b linked to the monoclonal anti-human CD105 (hCD105) antibody 44G4. The immunotoxin kills specifically mouse fibroblasts expressing the biomarker CD105 (L929-hCD105+ cells) with an IC(50) value of 6x10(-10)M while nigrin b does it at 2.4x10(-7)M. Immunofluorescence analysis indicated that the immunotoxin accumulates in a perinuclear region. In contrast, 44G4 showed a specific localization on the cell surface.
Collapse
Affiliation(s)
- Raquel Muñoz
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Valladolid, E-47005 Valladolid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Immunotoxins are proteins used to treat cancer that are composed of an antibody fragment linked to a toxin. The immunotoxin binds to a surface antigen on a cancer cell, enters the cell by endocytosis, and kills it. The most potent immunotoxins are made from bacterial and plant toxins. Refinements over many years have produced recombinant immunotoxins; these therapeutic proteins are made using protein engineering. Individual immunotoxins are designed to treat specific cancers. To date, most success has been achieved treating hematologic tumors. Obstacles to successful treatment of solid tumors include poor penetration into tumor masses and the immune response to the toxin component of the immunotoxin, which limits the number of cycles that can be given. Strategies to overcome these limitations are being pursued.
Collapse
Affiliation(s)
- Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | |
Collapse
|
25
|
Abstract
Objective clinical responses to anticancer treatments often do not translate into substantial improvements in overall survival. Recent data suggesting many cancers arise from rare self-renewing cells (cancer stem cells) that are biologically distinct from their more numerous differentiated progeny, may explain this paradox. Current anticancer therapies have been developed to target the bulk of the tumor mass (i.e., the differentiated cancer cells). Although treatments directed against the bulk of the cancer may produce dramatic responses, they are unlikely to result in long-term remissions if the rare cancer stem cells are also not targeted. Better understanding the biology of cancer stem cells as well reexamining both our preclinical and clinical drug development paradigms to include the cancer stem cell concept, have the potential to revolutionize the treatment of many cancers.
Collapse
|
26
|
Gökbuget N, Hoelzer D. Novel antibody-based therapy for acute lymphoblastic leukaemia. Best Pract Res Clin Haematol 2006; 19:701-13. [PMID: 16997178 DOI: 10.1016/j.beha.2006.06.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In recent decades rapid improvements in the results of treatment of adult acute lymphoblastic leukaemia (ALL) have been achieved. This progress has been based mainly on intensification and optimization of chemotherapy, risk-adapted use of stem-cell transplantation, and improved supportive care. However, results in adult patients are still considerably inferior to those in paediatric ALL, and a barrier to further intensification of chemotherapy appears to have been reached regarding toxicity. More recently, the most significant progress has therefore been achieved by individualized and targeted therapy - for example, treatment with monoclonal antibodies (MoAbs). ALL blast cells express a variety of specific antigens which may serve as targets: e.g. CD19, CD20, CD22, CD33, and CD52. Most experience is available for anti-CD20 (rituximab). In ALL, rituximab is combined with chemotherapy mainly in mature B-ALL and Burkitt's lymphoma, and interim results are very promising. Recently studies with rituximab have also been initiated in B-precursor ALL. Other antibodies would be of interest due to a high rate of antigen (e.g. CD19) expression in ALL, but these are not yet generally available. Clinical application in smaller studies or case reports was reported for anti-CD52 and anti-CD33. Overall it can be stated that MoAb therapy in ALL is a promising treatment approach. Monotherapy with MoAbs in relapsed ALL has also occasionally achieved responses, but greater effects can be expected from combination with chemotherapy and treatment in the state of minimal residual disease. Details of these regimens - required level of antigen expression, timing, schedule, dosage and stage of disease - remain to be defined.
Collapse
Affiliation(s)
- Nicola Gökbuget
- J W Goethe University Hospital, Medical Clinic III, Theodor Stern Kai 7, 60590 Frankfurt, Germany.
| | | |
Collapse
|
27
|
Abstract
Immunotoxins are proteins that contain a toxin along with an antibody or growth factor that binds specifically to target cells. Nearly all protein toxins work by enzymatically inhibiting protein synthesis. For the immunotoxin to work, it must bind to and be internalized by the target cells, and the enzymatic fragment of the toxin must translocate to the cytosol. Once in the cytosol, 1 molecule is capable of killing a cell, making immunotoxins some of the most potent killing agents. Various plant and bacterial toxins have been genetically fused or chemically conjugated to ligands that bind to cancer cells. Among the most active clinically are those that bind to hematologic tumors. At present, only 1 agent, which contains human interleukin-2 and truncated diphtheria toxin, is approved for use in cutaneous T-cell lymphoma. Another, containing an anti-CD22 Fv and truncated Pseudomonas exotoxin, has induced complete remissions in a high proportion of cases of hairy-cell leukemia. Refinement of existing immunotoxins and development of new immunotoxins are underway to improve the treatment of cancer.
Collapse
Affiliation(s)
- Robert J Kreitman
- Clinical Immunotherapy Section, Laboratory of Molecular Biology, Centers for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 37, Room 5124B, Bethesda, MD 20892-4255, USA.
| |
Collapse
|
28
|
Barbin K, Stieglmaier J, Saul D, Stieglmaier K, Stockmeyer B, Pfeiffer M, Lang P, Fey GH. Influence of variable N-glycosylation on the cytolytic potential of chimeric CD19 antibodies. J Immunother 2006; 29:122-33. [PMID: 16531813 DOI: 10.1097/01.cji.0000175684.28615.7b] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
To investigate the influence of N-linked oligosaccharides at asparagines-297 on the cytolytic potential of chimeric CD19 antibodies, three distinct variants were generated by production in different expression systems. The same chimeric CD19 antibody was produced in Sf21 insect cells, human 293 T cells, and 293 T cells expressing a co-transfected beta1,4-N-acetylglucosaminyltransferase III (GnTIII). The N-glycan structures and the cytolytic potential of the antibodies produced in these three systems were directly compared. After expression in insect cells, the antibody carried paucimannosidic N-linked oligosaccharides, distinct from the complex biantennary carbohydrate moieties attached to the product from human cells. After co-expression with GnTIII in human cells, the antibody carried an eightfold greater percentage of oligosaccharides with a bisecting N-acetylglucosamine (78.7% versus 9.6%) and a 30-fold increased proportion of bisecting, defucosylated oligosaccharides (15.9% versus 0.5%). The insect cell product triggered stronger antibody-dependent cellular cytotoxicity (ADCC) of a human leukemia-derived cell line than the product from non-re-engineered 293 T cells and was equally effective at 50- to 100-fold lower concentrations. The antibody from glyco-engineered 293 T cells had comparable lytic activity as the insect cell product. Both mediated significant ADCC at lower effector-to-target cell ratios than the antibody from non-re-engineered 293 T cells, and both were highly effective against primary blasts from pediatric leukemia patients. The data demonstrate the influence of the N-glycosylation pattern on the ADCC activity of chimeric CD19 antibodies and point to the importance of suitable expression systems for the production of highly active therapeutic antibodies.
Collapse
Affiliation(s)
- Karin Barbin
- Genetics, University of Erlangen--Nuremberg, D 91058 Erlangen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Huff CA, Matsui WH, Smith BD, Jones RJ. Strategies to eliminate cancer stem cells: clinical implications. Eur J Cancer 2006; 42:1293-7. [PMID: 16644203 DOI: 10.1016/j.ejca.2006.01.045] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2006] [Accepted: 01/23/2006] [Indexed: 12/31/2022]
Abstract
Over the past two decades, major advances in our understanding of cancer have translated into only modest increments in survival for the majority of cancer patients. Recent data suggesting cancers arise from rare self-renewing stem cells that are biologically distinct from their more numerous differentiated progeny may explain this paradox. Current anticancer therapies have been developed to decrease the bulk of the tumour mass (i.e. the differentiated cancer cells). Although treatments directed against the bulk of the cancer may produce dramatic responses, they are unlikely to result in long-term remissions if the rare cancer stem cells are also not targeted. Conversely, treatments that selectively attack cancer stem cells will not immediately eliminate the differentiated cancer cells, and might therefore be prematurely abandoned if clinical activity is judged solely by traditional response criteria that reflect changes in the bulk of the tumour. Re-examining both our pre-clinical and clinical drug development paradigms to include the cancer stem cell concept has the potential to revolutionize the treatment of many cancers.
Collapse
Affiliation(s)
- Carol Ann Huff
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Bunting-Blaustein Cancer Research Building, Baltimore, MD 21231, USA.
| | | | | | | |
Collapse
|
30
|
Abstract
Although most patients with adult acute lymphoblastic leukaemia (ALL) can achieve a remission when treated with conventional, DNA-damaging chemotherapy, in more than half of all cases the disease relapses and ultimately results in death. Therefore, there is a substantial need for new antileukaemic drugs. Recent advances in the understanding of the molecular alterations in ALL have lead to the identification of new targets and the arrival of molecular-targeted therapies in the clinical setting. The prototype for this approach is the treatment of Philadelphia chromosome-positive ALL with imatinib mesylate. Here, the targeting of a molecular abnormality--inhibition of BCR-ABL tyrosine kinase--has turned a very poor-prognosis disease into one in which promising results are achieved. Promising new therapies are under development that target various goals, including the NOTCH signalling pathway, purine nucleoside phosphorylase activity, mammalian target of rapamycin and tyrosine kinase. This review outlines recent advances in the development of emerging drugs for the treatment of adult ALL. The recent advances in the understanding of the biology and pathogenesis of ALL have helped to determine prognosis and to plan the therapy of adult patients with ALL. Still, despite improved complete remission rates of 65-90% with current therapy, only 20-40% of patients can be considered cured. New therapeutic alternatives are needed to improve these results. With a better understanding of the disease, more target-specific therapies could be designed. The aim of this review is to highlight new pharmacotherapies and those emerging drug treatments for patients with adult ALL.
Collapse
Affiliation(s)
- Xavier Thomas
- Department of Hematology, Edouard Herriot Hospital, Lyon, France.
| |
Collapse
|
31
|
Abstract
Immunophenotyping was introduced into diagnostic pathology over 30 years ago to assist in the diagnosis and classification of lymphoproliferative disorders. Today the role of immunophenotyping has been expanded beyond this to include the detection of markers of prognosis, determination of disease phenotypes associated with specific chromosomal abnormalities, detection of targets for immunotherapy and to monitor residual disease. Immunoperoxidase detection methods remain the most popular in histopathology, whilst flow cytometry is most commonly applied for haematological samples. The range of monoclonal antibodies available, including those which work in routinely performed tissue specimens, continues to increase. This is in part a result of gene expression studies identifying precise genetic signatures for certain lymphoproliferative disorders and the generation of new protein markers to gene products of upregulated genes. This review summarises the current status and applications of immunophenotyping in the assessment of many of the lymphoid malignancies.
Collapse
Affiliation(s)
- Emma J Gudgin
- Haematology Department, Addenbrooke's Hospital, Cambridge, United Kingdom
| | | |
Collapse
|
32
|
Szalai K, Schöll I, Förster-Waldl E, Polito L, Bolognesi A, Untersmayr E, Riemer AB, Boltz-Nitulescu G, Stirpe F, Jensen-Jarolim E. Occupational sensitization to ribosome-inactivating proteins in researchers. Clin Exp Allergy 2005; 35:1354-60. [PMID: 16238796 DOI: 10.1111/j.1365-2222.2005.02338.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Ribosome-inactivating proteins (RIPs) are expressed in many plants. Because of their anti-infectious and anti-proliferative effects, intensive research is going on for applying these toxins in therapy against viral infections or malignancies. Recently, we demonstrated that type I allergy against RIPs from elderberry can occur. OBJECTIVE Stimulated by our study, a group of RIP researchers reported that some of the employees had suspected allergy to RIPs. METHODS AND RESULTS We tested their sera in ELISA on natural RIPs. Specific IgE in four subjects were found against dianthin30, gelonin, momordin, PAP-S, saporin, ricin and volkensin. In contrast, asparin and lychnin did not show any IgE binding. When separating extracts of plants containing the toxins in SDS-PAGE, RIPs appeared to be the predominant constituents. Interestingly, among the other plant proteins, they were exclusively recognized by IgE in immunoblot. RIPs derived from close botanical families share high sequence homologies. Nevertheless, in IgE inhibition experiments with human sera, cross-reactivity between RIPs also derived from non-related plants could be demonstrated. CONCLUSION We conclude that sensitization and IgE induction to RIPs may occur upon exposure. This has to be considered when applying them in therapy against malignancies or viral infections.
Collapse
Affiliation(s)
- K Szalai
- Center of Physiology and Pathophysiology, University Children's Hospital, Medical University of Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Huff CA, Matsui W, Smith BD, Jones RJ. The paradox of response and survival in cancer therapeutics. Blood 2005; 107:431-4. [PMID: 16150939 PMCID: PMC1895602 DOI: 10.1182/blood-2005-06-2517] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Although most patients with cancer respond to therapy, few are cured. Moreover, objective clinical responses to treatment often do not even translate into substantial improvements in overall survival. For example, patients with indolent lymphoma who achieved a complete remission with conventional-dose therapies in the prerituximab era did not experience a survival advantage over similar patients treated with a "watch and wait" approach. Several studies have also shown that neither the magnitude nor the kinetics of clinical response has an impact on survival in multiple myeloma. Recent data suggesting many malignancies arise from a rare population of cells that exclusively maintains the ability to self-renew and sustains the tumor (ie, "cancer stem cells") may help explain this paradox that response and survival are not always linked. Therapies that successfully eliminate the differentiated cancer cells characterizing the tumor may be ineffective against rare, biologically distinct cancer stem cells. New methods for assessing treatment efficacy must also be developed, as traditional response criteria measure tumor bulk and may not reflect changes in rare cancer stem cell populations. In this article, we discuss the evidence for cancer stem cells in hematologic malignancies and possible ways to begin targeting these cells and measuring clinical effectiveness of such treatment approaches.
Collapse
Affiliation(s)
- Carol Ann Huff
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.
| | | | | | | |
Collapse
|
34
|
Parikh BA, Baykal U, Di R, Tumer NE. Evidence for retro-translocation of pokeweed antiviral protein from endoplasmic reticulum into cytosol and separation of its activity on ribosomes from its activity on capped RNA. Biochemistry 2005; 44:2478-90. [PMID: 15709760 DOI: 10.1021/bi048188c] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Pokeweed antiviral protein (PAP) is a single-chain ribosome inactivating protein (RIP) that binds to ribosomes and depurinates the highly conserved alpha-sarcin/ricin loop (SRL) of the large subunit rRNA. Catalytic depurination of a specific adenine has been proposed to result in translation arrest and cytotoxicity. Here, we show that both precursor and mature forms of PAP are localized in the endoplasmic reticulum (ER) in yeast. The mature form is retro-translocated from the ER into the cytosol where it escapes degradation unlike the other substrates of the retro-translocation pathway. A mutation of a highly conserved asparagine residue at position 70 (N70A) delays ribosome depurination and the onset of translation arrest. The ribosomes are eventually depurinated, yet cytotoxicity and loss of viability are markedly absent. Analysis of the variant protein, N70A, does not reveal any decrease in the rate of synthesis, subcellular localization, or the rate of transport into the cytosol. N70A destabilizes its own mRNA, binds to cap, and blocks cap dependent translation, as previously reported for the wild-type PAP. However, it cannot depurinate ribosomes in a translation-independent manner. These results demonstrate that N70 near the active-site pocket is required for depurination of cytosolic ribosomes but not for cap binding or mRNA destabilization, indicating that the activity of PAP on capped RNA can be uncoupled from its activity on rRNA. These findings suggest that the altered active site of PAP might accommodate a narrower range of substrates, thus reducing ribotoxicity while maintaining potential therapeutic benefits.
Collapse
Affiliation(s)
- Bijal A Parikh
- Graduate Program in Microbiology and Molecular Genetics, Rutgers University, New Brunswick, New Jersey 08901-8520, USA
| | | | | | | |
Collapse
|
35
|
Cooper LJN, Al-Kadhimi Z, DiGiusto D, Kalos M, Colcher D, Raubitschek A, Forman SJ, Jensen MC. Development and application of CD19-specific T cells for adoptive immunotherapy of B cell malignancies. Blood Cells Mol Dis 2005; 33:83-9. [PMID: 15223016 DOI: 10.1016/j.bcmd.2004.03.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2004] [Indexed: 11/27/2022]
Abstract
The graft-versus-leukemia (GVL)-effect achieved by donor-derived T cells arising from transplanted allogeneic hematopoietic stem cells or given as donor-leukocyte infusions (DLI) after allogeneic transplant, demonstrates that donor-derived T cells can eradicate B-lineage malignancies. However, graft-versus-host-disease (GVHD) occurring after allogeneic hematopoietic stem-cell transplant (HSCT) or polyclonal DLI can limit the efficacy of these interventions. This toxicity can be avoided by using autologous T cells and/or tumor-specific cytotoxic T lymphocytes (CTLs). To generate antigen-specific T cells that can be derived from the allogeneic donor or the patient, we have genetically manipulated T cells to express a CD19-specific chimeric immunoreceptor. This renders T cells specific for CD19, a cell surface molecule found on B-lineage leukemia and lymphoma. This review will demonstrate the redirected specificity of CD19-specific T cells and implementation of clinical trials using these cellular agents.
Collapse
Affiliation(s)
- Laurence J N Cooper
- Division of Hematology and Hematopoietic Cell Transplantation Transplant, Beckman Research Institute and City of Hope National Medical Center, Duarte, CA 91010-3000, USA.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Antolín P, Girotti A, Arias FJ, Barriuso B, Jiménez P, Rojo MA, Girbés T. Bacterial expression of biologically active recombinant musarmin 1 from bulbs of Muscari armeniacum L. and Miller. J Biotechnol 2004; 112:313-22. [PMID: 15313008 DOI: 10.1016/j.jbiotec.2004.04.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2003] [Revised: 04/27/2004] [Accepted: 04/30/2004] [Indexed: 10/26/2022]
Abstract
Musarmins are type 1 ribosome-inactivating proteins with N-glycosidase activity on the 28 S rRNA that are present in bulbs of Muscari armeniacum L. and Miller at rather low concentrations. In the present work, a cDNA fragment coding for musarmin 1 was sub-cloned and expressed in Escherichia coli. The recombinant protein (rMU1) was synthesised as a polypeptide of 295 amino acids that was delivered to the periplasm and processed. Recombinant musarmin 1 present in the periplam has two forms: insoluble with a molecular mass of 29,423 and soluble with a molecular mass of 29,117 because of a small proteolytic shortening with respect to the insoluble one, presumably in the C-terminal. The yield of protein homogeneous by polyacrylamide gel electrophoresis was 23mgl-1 of bacterial culture. The recombinant musarmin 1 forms isolated from both the soluble and the insoluble (upon refolding) fractions retained full translational inhibitory and 28 S rRNA N-glycosidase activities as compared with the native protein. The recombinant protein displayed great stability towards trypsin, collagenase, rat plasma and rat liver protein extract, but was sensitive to the action of papain and proteinase K. The easy availability and full activity of the recombinant musarmin 1 makes it a good candidate for the preparation of immunotoxins for targeted therapy and for the construction of transgenic plants expressing it as antipathogenic agent.
Collapse
Affiliation(s)
- Pilar Antolín
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Valladolid, 47005 Valladolid, Spain
| | | | | | | | | | | | | |
Collapse
|
37
|
Hudak KA, Parikh BA, Di R, Baricevic M, Santana M, Seskar M, Tumer NE. Generation of pokeweed antiviral protein mutations in Saccharomyces cerevisiae: evidence that ribosome depurination is not sufficient for cytotoxicity. Nucleic Acids Res 2004; 32:4244-56. [PMID: 15304562 PMCID: PMC514378 DOI: 10.1093/nar/gkh757] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Pokeweed antiviral protein (PAP) is a ribosome-inactivating protein that depurinates the highly conserved alpha-sarcin/ricin loop in the large rRNA. Here, using site-directed mutagenesis and systematic deletion analysis from the 5' and the 3' ends of the PAP cDNA, we identified the amino acids important for ribosome depurination and cytotoxicity of PAP. Truncating the first 16 amino acids of PAP eliminated its cytotoxicity and the ability to depurinate ribosomes. Ribosome depurination gradually decreased upon the sequential deletion of C-terminal amino acids and was abolished when a stop codon was introduced at Glu-244. Cytotoxicity of the C-terminal deletion mutants was lost before their ability to depurinate ribosomes. Mutations in Tyr-123 at the active site affected cytotoxicity without altering the ribosome depurination ability. Total translation was not inhibited in yeast expressing the non-toxic Tyr-123 mutants, although ribosomes were depurinated. These mutants depurinated ribosomes only during their translation and could not depurinate ribosomes in trans in a translation-independent manner. A mutation in Leu-71 in the central domain affected cytotoxicity without altering the ability to depurinate ribosomes in trans and inhibit translation. These results demonstrate that the ability to depurinate ribosomes in trans in a catalytic manner is required for the inhibition of translation, but is not sufficient for cytotoxicity.
Collapse
Affiliation(s)
- Katalin A Hudak
- Biotechnology Center for Agriculture and the Environment and the Department of Plant Biology and Pathology, Cook College, Rutgers University, New Brunswick, NJ 08901-8520, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Affiliation(s)
- Richard J Jones
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.
| | | | | |
Collapse
|
39
|
Wetzler M, Dodge RK, Mrózek K, Stewart CC, Carroll AJ, Tantravahi R, Vardiman JW, Larson RA, Bloomfield CD. Additional cytogenetic abnormalities in adults with Philadelphia chromosome-positive acute lymphoblastic leukaemia: a study of the Cancer and Leukaemia Group B. Br J Haematol 2004; 124:275-88. [PMID: 14717774 DOI: 10.1046/j.1365-2141.2003.04736.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We analysed the nature and prognostic significance of secondary cytogenetic changes in 111 newly diagnosed adults with acute lymphoblastic leukaemia (ALL) and t(9;22)(q34;q11.2) or its variants. Secondary aberrations were seen in 75 (68%) patients. They included, in order of descending frequency: +der(22)t(9;22), +21, abnormalities of 9p, high hyperdiploidy (>50 chromosomes), +8, -7, +X and abnormalities resulting in loss of material from 8p, gain of 8q, gain of 1q and loss of 7p. Eighty patients (72%) had > or =1 normal metaphase in their karyotype. There were four balanced and 12 unbalanced translocations previously unreported in ALL with t(9;22). The t(2;7)(p11;p13) and der(18)t(8;18)(q11.2;p11.2) were seen in two cases each, and have never before been reported in haematological malignancy. All but four patients were treated on front-line Cancer and Leukaemia Group B clinical protocols. The presence of -7 as a sole secondary abnormality was associated with a lower complete remission (CR) rate (P = 0.004), while the presence of > or =3 aberrations was associated with a higher CR rate (P = 0.009) and +der(22)t(9;22) with a higher cumulative incidence of relapse (P = 0.02). It will be of interest to see if newly diagnosed t(9;22)-positive adult ALL patients with these and other secondary aberrations respond differently to treatment regimens that include imatinib mesylate.
Collapse
Affiliation(s)
- Meir Wetzler
- Department of Medicine, Leukemia Section, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Recent publications in hematological oncology. Hematol Oncol 2003; 21:141-8. [PMID: 14594017 DOI: 10.1002/hon.708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|