1
|
Brock K, Homer V, Soul G, Potter C, Chiuzan C, Lee S. Is more better? An analysis of toxicity and response outcomes from dose-finding clinical trials in cancer. BMC Cancer 2021; 21:777. [PMID: 34225682 PMCID: PMC8256624 DOI: 10.1186/s12885-021-08440-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 06/04/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The overwhelming majority of dose-escalation clinical trials use methods that seek a maximum tolerable dose, including rule-based methods like the 3+3, and model-based methods like CRM and EWOC. These methods assume that the incidences of efficacy and toxicity always increase as dose is increased. This assumption is widely accepted with cytotoxic therapies. In recent decades, however, the search for novel cancer treatments has broadened, increasingly focusing on inhibitors and antibodies. The rationale that higher doses are always associated with superior efficacy is less clear for these types of therapies. METHODS We extracted dose-level efficacy and toxicity outcomes from 115 manuscripts reporting dose-finding clinical trials in cancer between 2008 and 2014. We analysed the outcomes from each manuscript using flexible non-linear regression models to investigate the evidence supporting the monotonic efficacy and toxicity assumptions. RESULTS We found that the monotonic toxicity assumption was well-supported across most treatment classes and disease areas. In contrast, we found very little evidence supporting the monotonic efficacy assumption. CONCLUSIONS Our conclusion is that dose-escalation trials routinely use methods whose assumptions are violated by the outcomes observed. As a consequence, dose-finding trials risk recommending unjustifiably high doses that may be harmful to patients. We recommend that trialists consider experimental designs that allow toxicity and efficacy outcomes to jointly determine the doses given to patients and recommended for further study.
Collapse
Affiliation(s)
- Kristian Brock
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK.
| | - Victoria Homer
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Gurjinder Soul
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Claire Potter
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Cody Chiuzan
- Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Shing Lee
- Mailman School of Public Health, Columbia University, New York, NY, USA
| |
Collapse
|
2
|
Silva RB, Yap C, Carvajal R, Lee SM. Would the Recommended Dose Have Been Different Using Novel Dose-Finding Designs? Comparing Dose-Finding Designs in Published Trials. JCO Precis Oncol 2021; 5:PO.21.00136. [PMID: 34250415 DOI: 10.1200/po.21.00136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/03/2021] [Accepted: 05/06/2021] [Indexed: 11/20/2022] Open
Abstract
Simulation studies have shown that novel designs such as the continual reassessment method and the Bayesian optimal interval (BOIN) design outperform the 3 + 3 design by recommending the maximum tolerated dose (MTD) more often, using less patients, and allotting more patients to the MTD. However, it is not clear whether these novel designs would have yielded different results in the context of real-world dose-finding trials. This is a commonly mentioned reason for the continuous use of 3 + 3 designs for oncology trials, with investigators considering simulation studies not sufficiently convincing to warrant the additional design complexity of novel designs. METHODS We randomly sampled 60 published dose-finding trials to obtain 22 that used the 3 + 3 design, identified an MTD, published toxicity data, and had more than two dose levels. We compared the published MTD with the estimated MTD using the continual reassessment method and BOIN using target toxicity rates of 25% and 30% and toxicity data from the trial. Moreover, we compared patient allocation and sample size assuming that these novel designs had been implemented. RESULTS Model-based designs chose dose levels higher than the published MTD in about 40% of the trials, with estimated and observed toxicity rates closer to the target toxicity rates of 25% and 30%. They also assigned less patients to suboptimal doses and permitted faster dose escalation. CONCLUSION This study using published dose-finding trials shows that novel designs would recommend different MTDs and confirms the advantages of these designs compared with the 3 + 3 design, which were demonstrated by simulation studies.
Collapse
Affiliation(s)
- Rebecca B Silva
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY
| | - Christina Yap
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Richard Carvajal
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY
| | - Shing M Lee
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY.,Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY
| |
Collapse
|
3
|
Kang L, Li C, Rosenkrans ZT, Huo N, Chen Z, Ehlerding EB, Huo Y, Ferreira CA, Barnhart TE, Engle JW, Wang R, Jiang D, Xu X, Cai W. CD38-Targeted Theranostics of Lymphoma with 89Zr/ 177Lu-Labeled Daratumumab. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2001879. [PMID: 34026426 PMCID: PMC8132161 DOI: 10.1002/advs.202001879] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 12/30/2020] [Indexed: 05/05/2023]
Abstract
Lymphoma is a heterogeneous disease with varying clinical manifestations and outcomes. Many subtypes of lymphoma, such as Burkitt's lymphoma and diffuse large B cell lymphoma, are highly aggressive with dismal prognosis even after conventional chemotherapy and radiotherapy. As such, exploring specific biomarkers for lymphoma is of high clinical significance. Herein, a potential marker, CD38, is investigated for differentiating lymphoma. A CD38-targeting monoclonal antibody (mAb, daratumumab) is then radiolabeled with Zr-89 and Lu-177 for theranostic applications. As the diagnostic component, the Zr-89-labeled mAb is highly specific in delineating CD38-positive lymphoma via positron emission tomography (PET) imaging, while the Lu-177-labeled mAb serves well as the therapeutic component to suppress tumor growth after a one-time administration. These results strongly suggest that CD38 is a lymphoma-specific marker and prove that 89Zr/177Lu-labeled daratumumab facilitates immunoPET imaging and radioimmunotherapy of lymphoma in preclinical models. Further clinical evaluation and translation of this CD38-targeted theranostics may be of significant help in lymphoma patient stratification and management.
Collapse
MESH Headings
- ADP-ribosyl Cyclase 1/immunology
- ADP-ribosyl Cyclase 1/metabolism
- Animals
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal/pharmacology
- Cell Line, Tumor
- Humans
- Immunologic Factors/pharmacokinetics
- Lutetium/pharmacokinetics
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/pathology
- Membrane Glycoproteins/immunology
- Membrane Glycoproteins/metabolism
- Mice, Inbred BALB C
- Mice, SCID
- Positron Emission Tomography Computed Tomography/methods
- Precision Medicine/methods
- Radioisotopes/pharmacokinetics
- Radiopharmaceuticals/pharmacokinetics
- Radiopharmaceuticals/pharmacology
- Tissue Distribution
- Xenograft Model Antitumor Assays
- Zirconium/pharmacokinetics
- Mice
Collapse
Affiliation(s)
- Lei Kang
- Department of Nuclear MedicinePeking University First HospitalBeijing100034China
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin – MadisonMadisonWI53705USA
| | - Cuicui Li
- Department of Nuclear MedicinePeking University First HospitalBeijing100034China
| | - Zachary T. Rosenkrans
- Department of Pharmaceutical SciencesUniversity of Wisconsin – MadisonMadisonWI53705USA
| | - Nan Huo
- Department of Medical Molecular BiologyBeijing Institute of BiotechnologyBeijing100850China
| | - Zhao Chen
- Department of Nuclear MedicinePeking University First HospitalBeijing100034China
| | - Emily B. Ehlerding
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin – MadisonMadisonWI53705USA
| | - Yan Huo
- Department of Nuclear MedicinePeking University First HospitalBeijing100034China
| | - Carolina A. Ferreira
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin – MadisonMadisonWI53705USA
| | - Todd E. Barnhart
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin – MadisonMadisonWI53705USA
| | - Jonathan W. Engle
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin – MadisonMadisonWI53705USA
| | - Rongfu Wang
- Department of Nuclear MedicinePeking University First HospitalBeijing100034China
| | - Dawei Jiang
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin – MadisonMadisonWI53705USA
- Department of Nuclear MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Xiaojie Xu
- Department of Medical Molecular BiologyBeijing Institute of BiotechnologyBeijing100850China
| | - Weibo Cai
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin – MadisonMadisonWI53705USA
- Department of Pharmaceutical SciencesUniversity of Wisconsin – MadisonMadisonWI53705USA
| |
Collapse
|
4
|
Allegra AG, Mannino F, Innao V, Musolino C, Allegra A. Radioprotective Agents and Enhancers Factors. Preventive and Therapeutic Strategies for Oxidative Induced Radiotherapy Damages in Hematological Malignancies. Antioxidants (Basel) 2020; 9:antiox9111116. [PMID: 33198328 PMCID: PMC7696711 DOI: 10.3390/antiox9111116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/08/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022] Open
Abstract
Radiation therapy plays a critical role in the management of a wide range of hematologic malignancies. It is well known that the post-irradiation damages both in the bone marrow and in other organs are the main causes of post-irradiation morbidity and mortality. Tumor control without producing extensive damage to the surrounding normal cells, through the use of radioprotectors, is of special clinical relevance in radiotherapy. An increasing amount of data is helping to clarify the role of oxidative stress in toxicity and therapy response. Radioprotective agents are substances that moderate the oxidative effects of radiation on healthy normal tissues while preserving the sensitivity to radiation damage in tumor cells. As well as the substances capable of carrying out a protective action against the oxidative damage caused by radiotherapy, other substances have been identified as possible enhancers of the radiotherapy and cytotoxic activity via an oxidative effect. The purpose of this review was to examine the data in the literature on the possible use of old and new substances to increase the efficacy of radiation treatment in hematological diseases and to reduce the harmful effects of the treatment.
Collapse
Affiliation(s)
- Andrea Gaetano Allegra
- Radiation Oncology Unit, Department of Biomedical, Experimental, and Clinical Sciences “Mario Serio”, Azienda Ospedaliero-Universitaria Careggi, University of Florence, 50100 Florence, Italy;
| | - Federica Mannino
- Department of Clinical and Experimental Medicine, University of Messina, c/o AOU Policlinico G. Martino, Via C. Valeria Gazzi, 98125 Messina, Italy;
| | - Vanessa Innao
- Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, Division of Haematology, University of Messina, 98125 Messina, Italy; (V.I.); (C.M.)
| | - Caterina Musolino
- Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, Division of Haematology, University of Messina, 98125 Messina, Italy; (V.I.); (C.M.)
| | - Alessandro Allegra
- Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, Division of Haematology, University of Messina, 98125 Messina, Italy; (V.I.); (C.M.)
- Correspondence: ; Tel.: +39-090-221-2364
| |
Collapse
|
5
|
Tafreshi NK, Doligalski ML, Tichacek CJ, Pandya DN, Budzevich MM, El-Haddad G, Khushalani NI, Moros EG, McLaughlin ML, Wadas TJ, Morse DL. Development of Targeted Alpha Particle Therapy for Solid Tumors. Molecules 2019; 24:molecules24234314. [PMID: 31779154 PMCID: PMC6930656 DOI: 10.3390/molecules24234314] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/11/2022] Open
Abstract
Targeted alpha-particle therapy (TAT) aims to selectively deliver radionuclides emitting α-particles (cytotoxic payload) to tumors by chelation to monoclonal antibodies, peptides or small molecules that recognize tumor-associated antigens or cell-surface receptors. Because of the high linear energy transfer (LET) and short range of alpha (α) particles in tissue, cancer cells can be significantly damaged while causing minimal toxicity to surrounding healthy cells. Recent clinical studies have demonstrated the remarkable efficacy of TAT in the treatment of metastatic, castration-resistant prostate cancer. In this comprehensive review, we discuss the current consensus regarding the properties of the α-particle-emitting radionuclides that are potentially relevant for use in the clinic; the TAT-mediated mechanisms responsible for cell death; the different classes of targeting moieties and radiometal chelators available for TAT development; current approaches to calculating radiation dosimetry for TATs; and lead optimization via medicinal chemistry to improve the TAT radiopharmaceutical properties. We have also summarized the use of TATs in pre-clinical and clinical studies to date.
Collapse
Affiliation(s)
- Narges K. Tafreshi
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (N.K.T.); (M.L.D.); (C.J.T.); (E.G.M.)
| | - Michael L. Doligalski
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (N.K.T.); (M.L.D.); (C.J.T.); (E.G.M.)
| | - Christopher J. Tichacek
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (N.K.T.); (M.L.D.); (C.J.T.); (E.G.M.)
| | - Darpan N. Pandya
- Department of Cancer Biology, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA; (D.N.P.); (T.J.W.)
| | - Mikalai M. Budzevich
- Small Animal Imaging Laboratory, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Ghassan El-Haddad
- Depts. of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Nikhil I. Khushalani
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Eduardo G. Moros
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (N.K.T.); (M.L.D.); (C.J.T.); (E.G.M.)
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
- Department of Physics, University of South Florida, Tampa, FL 33612, USA
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA
| | - Mark L. McLaughlin
- Department of Pharmaceutical Sciences, West Virginia University, Health Sciences Center, Morgantown, WV & Modulation Therapeutics Inc., 64 Medical Center Drive, Morgantown, WV 26506, USA;
| | - Thaddeus J. Wadas
- Department of Cancer Biology, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA; (D.N.P.); (T.J.W.)
| | - David L. Morse
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (N.K.T.); (M.L.D.); (C.J.T.); (E.G.M.)
- Department of Physics, University of South Florida, Tampa, FL 33612, USA
- Small Animal Imaging Laboratory, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA
- Correspondence: ; Tel.: +1-813-745-8948; Fax: +1-813-745-8375
| |
Collapse
|
6
|
Li HK, Morokoshi Y, Daino K, Furukawa T, Kamada T, Saga T, Hasegawa S. Transcriptomic Signatures of Auger Electron Radioimmunotherapy Using Nuclear Targeting (111)In-Trastuzumab for Potential Combination Therapies. Cancer Biother Radiopharm 2016; 30:349-58. [PMID: 26447839 DOI: 10.1089/cbr.2015.1882] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
(111)In-labeled trastuzumab modified with nuclear localizing signal (NLS) peptides ((111)In-trastuzumab-NLS) efficiently delivers an Auger electron (AE) emitter (111)In into the cell nucleus and is thus a promising radiopharmaceutical in AE radioimmunotherapy (AE-RIT) for targeted killing of HER2-positive cancer. However, further improvement of its therapeutic efficacy is required. In this study, the authors show a transcriptomic approach to identify potential targets for enhancing the cytotoxic effects of (111)In-trastuzumab-NLS. They generated two types of (111)In-trastuzumab-NLS harboring different numbers of NLS peptides, (111)In-trastuzumab-NLS-S and -L. These radioimmunoconjugates (230 and 460 kBq) showed a significant higher cytotoxicity to SKBR3 human breast cancer cells overexpressing HER2 compared to (111)In-trastuzumab. Microarray analysis revealed that NF-kB-related genes (38 genes) were significantly changed in transcription by (111)In trastuzumab-NLS-L (230 kBq) treatment. Quantitative reverse transcription polymerase chain reaction confirmed the microarray data by showing transcriptional alternation of selected NF-κB target genes in cells treated with (111)In-trastuzumab-NLS-L. Interestingly, bortezomib, a drug known as a NF-κB modulator, significantly enhanced the cytotoxicity of (111)In-trastuzumab-NLS-L in SKBR3 cells. Taken together, the transcriptome data suggest the possibility that the modulation of NF-kB signaling activity is a molecular signature of (111)In-trastuzumab-NLS and coadministration of bortezomib may be efficacious in enhancement of AE-RIT with (111)In-trastuzumab-NLS.
Collapse
Affiliation(s)
- Huizi Keiko Li
- 1 Molecular Imaging Center, National Institute of Radiological Sciences , Chiba, Japan .,2 Graduate School of Medical and Pharmaceutical Sciences, Chiba University , Chiba, Japan .,3 Research Center for Charged Particle Therapy, National Institute of Radiological Sciences, Chiba, Japan
| | - Yukie Morokoshi
- 1 Molecular Imaging Center, National Institute of Radiological Sciences , Chiba, Japan
| | - Kazuhiro Daino
- 4 Research Center for Radiation Protection, National Institute of Radiological Sciences, Chiba, Japan
| | - Takako Furukawa
- 1 Molecular Imaging Center, National Institute of Radiological Sciences , Chiba, Japan
| | - Tadashi Kamada
- 2 Graduate School of Medical and Pharmaceutical Sciences, Chiba University , Chiba, Japan .,3 Research Center for Charged Particle Therapy, National Institute of Radiological Sciences, Chiba, Japan
| | - Tsuneo Saga
- 1 Molecular Imaging Center, National Institute of Radiological Sciences , Chiba, Japan
| | - Sumitaka Hasegawa
- 1 Molecular Imaging Center, National Institute of Radiological Sciences , Chiba, Japan
| |
Collapse
|
7
|
Sun Z, Braun TM. A two-dimensional biased coin design for dual-agent dose-finding trials. Clin Trials 2015; 12:596-607. [DOI: 10.1177/1740774515592404] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Given the limited efficacy observed with single agents, there is growing interest in Phase I clinical trial designs that allow for identification of the maximum tolerated combination of two agents. Purpose: Existing parametric designs may suffer from over- or under-parameterization. Thus, we have designed a nonparametric approach that can be easily understood and implemented for combination trials. Methods: We propose a two-stage adaptive biased coin design that extends existing methods for single-agent trials to dual-agent dose-finding trials. The basic idea of our design is to divide the entire trial into two stages and apply the biased coin design, with modification, in each stage. Results: We compare the operating characteristics of our design to four competing parametric approaches via simulation in several numerical examples. Under all simulation scenarios we have examined, our method performs well in terms of identification of the maximum tolerated combination and allocation of patients relative to the performance of its competitors. Limitations: In our design, stopping rule criteria and the distribution of the total sample size among the two stages are context-dependent, and both need careful consideration before adopting our design in practice. Efficacy is not a part of the dose-assignment algorithm, nor used to define the maximum tolerated combination. Conclusion: Our design inherits the favorable statistical properties of the biased coin design, is competitive with existing designs, and promotes patient safety by limiting patient exposure to toxic combinations whenever possible.
Collapse
Affiliation(s)
- Zhichao Sun
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Thomas M Braun
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
8
|
Humm JL, Sartor O, Parker C, Bruland OS, Macklis R. Radium-223 in the treatment of osteoblastic metastases: a critical clinical review. Int J Radiat Oncol Biol Phys 2015; 91:898-906. [PMID: 25832684 DOI: 10.1016/j.ijrobp.2014.12.061] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 12/30/2014] [Accepted: 12/31/2014] [Indexed: 01/06/2023]
Abstract
The element radium (Ra) was discovered by the Curies in 1898 and within a decade was in broad scientific testing for the management of several forms of cancer. The compound was known to give rise to a series of both high-energy particulate and penetrating γ-emissions. The latter found an important role in early 20th century brachytherapy applications, but the short-range α-particles seemed much less useful. Although highly cytotoxic when released within a few cell diameters of critical cell nuclei, the dense double-strand break damage was poorly repaired, and concerns regarding treatment-related toxicities and secondary malignancies halted clinical development. Moreover, the most common isotope of Ra has an exceptionally long half-life (>1600 years for (226)Ra) that proved daunting when aiming for a systemic cancer therapy. Fortunately, other radium isotopes have more convenient half-lives while still producing cytotoxic α particles. Radium-223 dichloride has a half-life of 11.4 days, and this isotope was identified as an excellent candidate for radionuclide therapy of cancers metastatic to bone. The calcium-mimetic chemical properties of the radium allowed intravenous infusion with rapid uptake to sites of new bone formation. The highly efficient bone localization suggested a potential therapeutic role for osteoblastic bone metastases, and a series of phase 1, 2, and 3 clinical trials was undertaken to explore this possibility. This series of clinical explorations culminated in the ALSYMPCA trial, an international, placebo-controlled, phase 3 study that accrued 921 symptomatic men with bone-metastatic, castrate-resistant prostate cancer. Results of this trial demonstrated a prolongation of overall survival, and regulatory agencies around the world have now approved this product as a treatment for advanced prostate cancer.
Collapse
Affiliation(s)
- John L Humm
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Oliver Sartor
- Departments of Medicine and Urology, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana
| | - Chris Parker
- Department of Clinical Oncology, Institute of Cancer Research, Sutton, United Kingdom
| | - Oyvind S Bruland
- Department of Oncology, Norwegian Radium Hospital and Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Roger Macklis
- Department of Radiation Oncology, University Hospitals of Cleveland and Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|
9
|
Pacilio M, Ventroni G, Basile C, Ialongo P, Becci D, Mango L. Improving the dose-myelotoxicity correlation in radiometabolic therapy of bone metastases with 153Sm-EDTMP. Eur J Nucl Med Mol Imaging 2014; 41:238-52. [PMID: 24077786 DOI: 10.1007/s00259-013-2552-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 08/15/2013] [Indexed: 12/11/2022]
Abstract
PURPOSE (153)Sm-ethylene diamine tetramethylene phosphonic acid ((153)Sm-EDTMP) is widely used to palliate pain from bone metastases, and is being studied for combination therapy beyond palliation. Conceptually, red marrow (RM) dosimetry allows myelotoxicity to be predicted, but the correlation is poor due to dosimetric uncertainty, individual sensitivity and biological effects from previous treatments. According to EANM guidelines, basic dosimetric procedures have been studied to improve the correlation between dosimetry and myelotoxicity in (153)Sm-EDTMP therapy. METHODS RM dosimetry for 33 treatments of bone metastases from breast, prostate and lung tumours was performed prospectively (with (99m)Tc-MDP) and retrospectively, acquiring whole-body scans early and late after injection. The (153)Sm-EDTMP activity was calculated by prospective dosimetry based on measured skeletal uptake and full physical retention, with the RM absorbed dose not exceeding 3.8 Gy. Patient-specific RM mass was evaluated by scaling in terms of body weight (BW), lean body mass (LBM) and trabecular volume (TV) estimated from CT scans of the L2–L4 vertebrae. Correlations with toxicity were determined in a selected subgroup of 27 patients, in which a better correlation between dosimetry and myelotoxicity was expected. RESULTS Skeletal uptakes of (99m)Tc and (153)Sm (Tc% and Sm%) were well correlated. The median Sm% was higher in prostate cancer (75.3 %) than in lung (60.5%, p = 0.005) or breast (60.8%, p = 0.008). PLT and WBC nadirs were not correlated with administered activity, but were weakly correlated with uncorrected RM absorbed doses, and the correlation improved after rescaling in terms of BW, LBM and TV. Most patients showed transient toxicity (grade 1–3), which completely and spontaneously recovered over a few days. Using TV, RM absorbed dose was in the range 2–5 Gy, with a median of 312 cGy for PLT in patients with toxicity and 247 cGy in those with no toxicity (p = 0.019), and 312 cGy for WBC in those with toxicity and 232 cGy in those with no toxicity (p = 0.019). ROC curves confirmed the correlations, yielding toxicity absorbed dose thresholds of 265 cGy for PLT and 232 cGy for WBC. CONCLUSION The best predictor of myelotoxicity and blood cells nadir was obtained scaling the RM absorbed dose in terms of the estimated TV. It seems clear that the increase in skeletal uptake due to the presence of bone metastases and the assumption of full physical retention cause an overestimation of the RM absorbed dose. Nevertheless, an improvement of the dose–toxicity correlation is easily achievable by simple methods, also leading to possible improvement in multifactorial analyses of myelotoxicity.
Collapse
|
10
|
Green DJ, Orgun NN, Jones JC, Hylarides MD, Pagel JM, Hamlin DK, Wilbur DS, Lin Y, Fisher DR, Kenoyer AL, Frayo SL, Gopal AK, Orozco JJ, Gooley TA, Wood BL, Bensinger WI, Press OW. A preclinical model of CD38-pretargeted radioimmunotherapy for plasma cell malignancies. Cancer Res 2013; 74:1179-89. [PMID: 24371230 DOI: 10.1158/0008-5472.can-13-1589] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The vast majority of patients with plasma cell neoplasms die of progressive disease despite high response rates to novel agents. Malignant plasma cells are very radiosensitive, but the potential role of radioimmunotherapy (RIT) in the management of plasmacytomas and multiple myeloma has undergone only limited evaluation. Furthermore, CD38 has not been explored as a RIT target despite its uniform high expression on malignant plasma cells. In this report, both conventional RIT (directly radiolabeled antibody) and streptavidin-biotin pretargeted RIT (PRIT) directed against the CD38 antigen were assessed as approaches to deliver radiation doses sufficient for multiple myeloma cell eradication. PRIT demonstrated biodistributions that were markedly superior to conventional RIT. Tumor-to-blood ratios as high as 638:1 were seen 24 hours after PRIT, whereas ratios never exceeded 1:1 with conventional RIT. (90)Yttrium absorbed dose estimates demonstrated excellent target-to-normal organ ratios (6:1 for the kidney, lung, liver; 10:1 for the whole body). Objective remissions were observed within 7 days in 100% of the mice treated with doses ranging from 800 to 1,200 μCi of anti-CD38 pretargeted (90)Y-DOTA-biotin, including 100% complete remissions (no detectable tumor in treated mice compared with tumors that were 2,982% ± 2,834% of initial tumor volume in control animals) by day 23. Furthermore, 100% of animals bearing NCI-H929 multiple myeloma tumor xenografts treated with 800 μCi of anti-CD38 pretargeted (90)Y-DOTA-biotin achieved long-term myeloma-free survival (>70 days) compared with none (0%) of the control animals.
Collapse
Affiliation(s)
- Damian J Green
- Authors' Affiliations: Clinical Research Division, Fred Hutchinson Cancer Research Center; Departments of Medicine, Radiation Oncology, and Laboratory Medicine, University of Washington, Seattle; and Dade Moeller Health Group, Richland, Washington
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Rae C, Tesson M, Babich JW, Boyd M, Mairs RJ. Radiosensitization of noradrenaline transporter-expressing tumour cells by proteasome inhibitors and the role of reactive oxygen species. EJNMMI Res 2013; 3:73. [PMID: 24219987 PMCID: PMC3828419 DOI: 10.1186/2191-219x-3-73] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 10/01/2013] [Indexed: 12/24/2022] Open
Abstract
Background The radiopharmaceutical 131I-metaiodobenzylguanidine (131I-MIBG) is used for the targeted radiotherapy of noradrenaline transporter (NAT)-expressing neuroblastoma. Enhancement of 131I-MIBG's efficacy is achieved by combination with the topoisomerase I inhibitor topotecan - currently being evaluated clinically. Proteasome activity affords resistance of tumour cells to radiation and topoisomerase inhibitors. Therefore, the proteasome inhibitor bortezomib was evaluated with respect to its cytotoxic potency as a single agent and in combination with 131I-MIBG and topotecan. Since elevated levels of reactive oxygen species (ROS) are induced by bortezomib, the role of ROS in tumour cell kill was determined following treatment with bortezomib or the alternative proteasome inhibitor, MG132. Methods Clonogenic assay and growth of tumour xenografts were used to investigate the effects of proteasome inhibitors alone or in combination with radiation treatment. Synergistic interactions in vitro were evaluated by combination index analysis. The dependency of proteasome inhibitor-induced clonogenic kill on ROS generation was assessed using antioxidants. Results Bortezomib, in the dose range 1 to 30 nM, decreased clonogenic survival of both SK-N-BE(2c) and UVW/NAT cells, and this was prevented by antioxidants. It also acted as a sensitizer in vitro when administered with X-radiation, with 131I-MIBG, or with 131I-MIBG and topotecan. Moreover, bortezomib enhanced the delay of the growth of human tumour xenografts in athymic mice when administered in combination with 131I-MIBG and topotecan. MG132 and bortezomib had similar radiosensitizing potency, but only bortezomib-induced cytotoxicity was ROS-dependent. Conclusions Proteasome inhibition shows promise for the treatment of neuroblastoma in combination with 131I-MIBG and topotecan. Since the cytotoxicity of MG132, unlike that of bortezomib, was not ROS-dependent, the latter proteasome inhibitor may have a favourable toxicity profile in normal tissues.
Collapse
Affiliation(s)
| | | | | | | | - Robert J Mairs
- Radiation Oncology, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow G61 1BD, Scotland.
| |
Collapse
|
12
|
Wages NA, Conaway MR. Specifications of a continual reassessment method design for phase I trials of combined drugs. Pharm Stat 2013; 12:217-24. [PMID: 23729323 DOI: 10.1002/pst.1575] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In studies of combinations of agents in phase I oncology trials, the dose-toxicity relationship may not be monotone for all combinations, in which case the toxicity probabilities follow a partial order. The continual reassessment method for partial orders (PO-CRM) is a design for phase I trials of combinations that leans upon identifying possible complete orders associated with the partial order. This article addresses some practical design considerations not previously undertaken when describing the PO-CRM. We describe an approach in choosing a proper subset of possible orderings, formulated according to the known toxicity relationships within a matrix of combination therapies. Other design issues, such as working model selection and stopping rules, are also discussed. We demonstrate the practical ability of PO-CRM as a phase I design for combinations through its use in a recent trial designed at the University of Virginia Cancer Center.
Collapse
Affiliation(s)
- Nolan A Wages
- Division of Translational Research and Applied Statistics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA.
| | | |
Collapse
|
13
|
Shi Y, Yin G. Escalation with overdose control for phase I drug-combination trials. Stat Med 2013; 32:4400-12. [PMID: 23630103 DOI: 10.1002/sim.5832] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 04/02/2013] [Indexed: 11/06/2022]
Abstract
Dose finding for combined drugs has grown rapidly in oncology drug development. The escalation with overdose control (EWOC) method is a popular model-based dose-finding approach to single-agent phase I clinical trials. When two drugs are combined as a treatment, we propose a two-dimensional EWOC design for dose finding on the basis of a four-parameter logistic regression model. During trial conduct, we continuously update the posterior distribution of the maximum tolerated dose (MTD) combination to find the most appropriate dose combination for each cohort of patients. The probability that the next assigned dose combination exceeds the MTD combination can be controlled by a feasibility bound, which is based on a prespecified quantile level of the MTD distribution such as to reduce the possibility of overdosing. We determine dose escalation, de-escalation, or staying at the same doses by searching the MTD combination along the rows and columns in a two-drug combination matrix, respectively. We conduct simulation studies to examine the performance of the two-dimensional EWOC design under various practical scenarios, and illustrate it with a trial example.
Collapse
Affiliation(s)
- Yun Shi
- Department of Statistics and Actuarial Science, The University of Hong Kong, Pokfulam Road, Hong Kong
| | | |
Collapse
|
14
|
Sartor O, Hoskin P, Bruland ØS. Targeted radio-nuclide therapy of skeletal metastases. Cancer Treat Rev 2013; 39:18-26. [DOI: 10.1016/j.ctrv.2012.03.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 03/14/2012] [Accepted: 03/16/2012] [Indexed: 01/29/2023]
|
15
|
Abstract
Primary and metastatic malignant bone lesions result in significant pain and disability in oncology patients. Targeted bone-seeking radioisotopes including 153Samarium ethylene-diamine-tetramethylene-phosphonic acid (153Sm-EDTMP) have been shown to effectively palliate bone pain, often when external beam radiotherapy (EBRT) is not feasible. However, recent evidence also suggests 153Sm-EDTMP has cytotoxic activity either alone or in combination with chemotherapy or EBRT. 153Sm-EDTMP may be useful as anti-neoplastic therapy apart from pain palliation in a variety of malignancies. For prostate cancer patients, several phase I and II clinical trials have shown that combined 153Sm-EDTMP and docetaxel-based chemotherapy can result in >50% decrease in prostate-specific antigen with manageable myelosuppression. In hematologic malignancies, 153Sm-EDTMP produced clinical responses when combined with bortezomib in multiple myeloma. 153Sm-EDTMP also can be used with myeloablative chemotherapy for marrow conditioning prior to stem cell transplant. In osteosarcoma, 153Sm-EDTMP infusion delivers radiation to multiple unresectable lesions simultaneously and provides local cytotoxicity without soft tissue damage that can be combined with chemotherapy or radiation. Prior to routine incorporation of 153Sm-EDTMP into therapeutic regimens, we must learn how to ensure optimal delivery to tumors, determine which patients are likely to benefit, improve our ability to assess clinical response in bone lesions and further evaluate the efficacy 153Sm-EDTMP in combination with chemotherapy, radiation and novel targeted agents.
Collapse
Affiliation(s)
- Breelyn A Wilky
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - David M Loeb
- Department of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
16
|
Goel A, Spitz DR, Weiner GJ. Manipulation of cellular redox parameters for improving therapeutic responses in B-cell lymphoma and multiple myeloma. J Cell Biochem 2012; 113:419-25. [PMID: 21956712 DOI: 10.1002/jcb.23387] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Developing novel combined-modality therapeutic approaches based on understanding of the involvement of redox biology in apoptosis of malignant cells is a promising approach for improving clinical responses in B-cell lymphoma and multiple myeloma. Therapeutic modalities that generate reactive oxygen species (i.e., radiation, photodynamic therapy, and specific chemotherapeutic drugs) have been shown to be selectively cytotoxic to malignant B-cells. In this review, we will discuss agents that induce apoptosis in B-cell tumors by oxidative stress. Subsequently, a novel biochemical rationale (based on fundamental differences in cancer vs. normal cell oxidative metabolism) for combining oxidative stressors with radiotherapy and chemotherapy, that may lead to designing of more effective treatment strategies for B-cell malignancies, will be discussed. Besides providing potential curative benefit, such novel therapies could also selectively target and inhibit the emergence of drug-resistance in tumor cells, which is a major determinant of treatment failure in many B-cell malignancies.
Collapse
Affiliation(s)
- Apollina Goel
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, The Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52242, USA.
| | | | | |
Collapse
|
17
|
Abstract
In May 2003, the US Food and Drug Administration (FDA) granted accelerated approval for the use of the first-in-class proteasome inhibitor bortezomib as a third-line therapy in multiple myeloma, and the European Union followed suit a year later. Bortezomib has subsequently been approved for multiple myeloma as a second-line treatment on its own and as a first-line therapy in combination with an alkylating agent and a corticosteroid. Furthermore, bortezomib has also been approved as a second-line therapy for mantle cell lymphoma. In this chapter, the focus is on the current clinical research on bortezomib, its adverse effects, and the resistance of multiple myeloma patients to bortezomib-based therapy. The various applications of bortezomib in different diseases and recent advances in the development of a new generation of inhibitors that target the proteasome or other parts of the ubiquitin-proteasome system are also reviewed.
Collapse
Affiliation(s)
- Boris Cvek
- Department of Cell Biology & Genetics, Palacky University, Olomouc, Czech Republic
| |
Collapse
|
18
|
Chen RA, Tu Y, Cao Y, Liu L, Liang Y. Bortezomib-Dexamethasone or Vincristine-Doxorubicin-Dexamethasone as Induction Therapy Followed by Thalidomide as Maintenance Therapy in Untreated Multiple Myeloma Patients. J Int Med Res 2011; 39:1975-84. [PMID: 22118002 DOI: 10.1177/147323001103900544] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
This randomized, retrospective study evaluated the effect of thalidomide combined with bortezomib-dexamethasone (TBD) or vincristine-doxorubicin-dexamethasone (T-VAD) on 46 patients with multiple myeloma. Newly diagnosed patients were randomly allocated to receive TBD ( n = 24) or T-VAD ( n = 22). The high-quality response rate (complete response plus very good partial response) was 62.5% in the TBD group and 45.4% for T-VAD. The complete response rate was 29.2% for TBD and 13.6% for T-VAD. Overall survival at 2 and 3 years, respectively, was 91.7% and 62.5% for TBD versus 86.4% and 54.5% for T-VAD. Most of the toxic effects of treatment were well tolerated. Both regimens were effective in the treatment of newly diagnosed multiple myeloma patients. Further studies are required to determine the role of thalidomide in these two regimens.
Collapse
Affiliation(s)
- RA Chen
- Department of Haematology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Y Tu
- Department of Emergency, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Y Cao
- Department of Emergency, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - L Liu
- Department of Haematology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Y Liang
- Department of Haematology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
19
|
Waterman GN, Yellin O, Jamshidinia K, Swift RA, Tamkin JA, Audell RA, Berenson JR. Metatarsal stress fractures in patients with multiple myeloma treated with long-term bisphosphonates: a report of six cases. J Bone Joint Surg Am 2011; 93:e106. [PMID: 21938357 DOI: 10.2106/jbjs.j.00455] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Gabriel N Waterman
- Inc., 9201 West Sunset Boulevard, Suite 310, West Hollywood, CA 90069, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Dexamethasone-induced oxidative stress enhances myeloma cell radiosensitization while sparing normal bone marrow hematopoiesis. Neoplasia 2011; 12:980-92. [PMID: 21170263 DOI: 10.1593/neo.101146] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Revised: 09/15/2010] [Accepted: 09/17/2010] [Indexed: 12/20/2022] Open
Abstract
Dexamethasone (Dex) and radiation therapy are established modalities in multiple myeloma. In this study, we propose a novel combination of Dex plus radiation that shows superior clonogenic cell killing and apoptosis of myeloma cells and selectively eliminates myeloma cells when cocultured with bone marrow stromal cells (BMSCs). Dex was found to inhibit the release of interleukin-6 from irradiated BMSCs, which is an established myeloma cell proproliferative cytokine. In 5TGM1 model, the combination of Dex with skeletal targeted radiotherapy (153-Sm-EDTMP) prolonged median survival time and inhibited radiation-induced myelosuppression. A two-cycle treatment of Dex plus 153-Sm-EDTMP was well tolerated and further improved median survival time. Mechanistically, Dex increased superoxide and hydrogen peroxide production and augmented radiation-induced oxidative stress and cell death of myeloma cells. In contrast, Dex inhibited radiation-induced increase in pro-oxidant levels and enhanced the clonogenic survival in normal hematopoietic stem and progenitor cells. Treatment with either N-acetylcysteine or the combination of polyethylene glycol (PEG)-conjugated copper, zinc-superoxide dismutase, and PEG-catalase significantly protected myeloma cells from Dex-induced clonogenic death. Overall, these results demonstrate that Dex in combination with radiotherapy enhances the killing of myeloma cells while protecting normal bone marrow hematopoiesis through a mechanism that involves selective increases in oxidative stress.
Collapse
|
21
|
A Phase I Trial of Samarium-153-Lexidronam Complex for Treatment of Clinically Nonmetastatic High-Risk Prostate Cancer: First Report of a Completed Study. Int J Radiat Oncol Biol Phys 2011; 79:732-7. [DOI: 10.1016/j.ijrobp.2009.11.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Revised: 11/10/2009] [Accepted: 11/16/2009] [Indexed: 11/19/2022]
|
22
|
Abstract
Much of the statistical methodology underlying the experimental design of phase 1 trials in oncology is intended for studies involving a single cytotoxic agent. The goal of these studies is to estimate the maximally tolerated dose, the highest dose that can be administered with an acceptable level of toxicity. A fundamental assumption of these methods is monotonicity of the dose-toxicity curve. This is a reasonable assumption for single-agent trials in which the administration of greater doses of the agent can be expected to produce dose-limiting toxicities in increasing proportions of patients. When studying multiple agents, the assumption may not hold because the ordering of the toxicity probabilities could possibly be unknown for several of the available drug combinations. At the same time, some of the orderings are known and so we describe the whole situation as that of a partial ordering. In this article, we propose a new two-dimensional dose-finding method for multiple-agent trials that simplifies to the continual reassessment method (CRM), introduced by O'Quigley, Pepe, and Fisher (1990, Biometrics 46, 33-48), when the ordering is fully known. This design enables us to relax the assumption of a monotonic dose-toxicity curve. We compare our approach and some simulation results to a CRM design in which the ordering is known as well as to other suggestions for partial orders.
Collapse
Affiliation(s)
- Nolan A Wages
- Department of Mathematics and Computer Science, Hampden-Sydney College, Hampden-Sydney, Virginia 23943, USA.
| | | | | |
Collapse
|
23
|
Barker CA, Powell SN. Enhancing radiotherapy through a greater understanding of homologous recombination. Semin Radiat Oncol 2011; 20:267-273.e3. [PMID: 20832019 DOI: 10.1016/j.semradonc.2010.05.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Radiotherapy for the treatment of cancer can cause a wide range of cellular effects, the most biologically potent of which is the double-strand break in DNA. The process of repairing DNA double-strand breaks involves 1 of 2 major mechanisms: nonhomologous end joining or homologous recombination. In this review, we review the molecular mechanisms of homologous recombination, in particular as it relates to the repair of DNA damage from ionizing radiation. We also present specific situations in which homologous recombination may be dysfunctional in human cancers and how this functional abnormality can be recognized. We also discuss the therapeutic opportunities that can be exploited based on deficiencies in homologous recombination at various steps in the DNA repair pathway. Side-by-side with these potential therapeutic opportunities, we review the contemporary clinical trials in which strategies to exploit these defects in homologous recombination can be enhanced by the use of radiotherapy in conjunction with biologically targeted agents. We conclude that the field of radiation oncology has only scratched the surface of a potentially highly efficacious therapeutic strategy.
Collapse
Affiliation(s)
- Christopher A Barker
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
24
|
Abstract
Advanced multiple myeloma is typically accompanied by osteolytic bone lesions resulting from heightened osteolytic activity of osteoclasts and decreased rates of osteogenesis by osteoblasts. Therefore, patients with myeloma bone disease are at increased risk for skeletal-related events (SREs) such as pathologic fracture, the need for radiotherapy or surgery to bone, spinal cord compression, and hypercalcemia of malignancy. Each of these can reduce patients' functional independence, quality of life, and survival. Radiotherapy and surgery are often used to palliate bone pain and to stabilize, repair, or prevent bone fractures. Bisphosphonates (BPs) may reduce the risk of SREs. In particular, clodronate, pamidronate, and zoledronic acid (ZOL) have demonstrated efficacy for delaying the onset of potentially life-threatening SREs. Overall, BPs have a well established tolerability profile. The introduction of BPs for multiple myeloma was practice-changing, and patients now experience far fewer serious fractures and hypercalcemia of malignancy. Ongoing studies will help further refine and optimize the timing and duration of BP therapy in patients with myeloma bone disease.
Collapse
Affiliation(s)
- James R Berenson
- Institute for Myeloma and Bone Cancer Research, West Hollywood, CA, USA.
| |
Collapse
|
25
|
Shah JJ, Orlowski RZ. Proteasome inhibitors in the treatment of multiple myeloma. Leukemia 2009; 23:1964-79. [PMID: 19741722 PMCID: PMC4737506 DOI: 10.1038/leu.2009.173] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Revised: 06/16/2009] [Accepted: 06/29/2009] [Indexed: 01/09/2023]
Abstract
Targeting intracellular protein turnover by inhibiting the ubiquitin-proteasome pathway as a strategy for cancer therapy is a new addition to our chemotherapeutic armamentarium, and has seen its greatest successes against multiple myeloma. The first-in-class proteasome inhibitor, bortezomib, was initially approved for treatment of patients in the relapsed/refractory setting as a single agent, and was recently shown to induce even greater benefits as part of rationally designed combinations that overcome chemoresistance. Modulation of proteasome function is also a rational approach to achieve chemosensitization to other antimyeloma agents, and bortezomib has now been incorporated into the front-line setting. Bortezomib-based induction regimens are able to achieve higher overall response rates and response qualities than was the case with prior standards of care, and unlike these older approaches, maintain efficacy in patients with clinically and molecularly defined high-risk disease. Second-generation proteasome inhibitors with novel properties, such as NPI-0052 and carfilzomib, are entering the clinical arena, and showing evidence of antimyeloma activity. In this spotlight review, we provide an overview of the current state of the art use of bortezomib and other proteasome inhibitors against multiple myeloma, and highlight areas for future study that will further optimize our ability to benefit patients with this disease.
Collapse
Affiliation(s)
- Jatin J. Shah
- The University of Texas M. D. Anderson Cancer Center, Department of Lymphoma & Myeloma, Houston, TX
| | - Robert Z. Orlowski
- The University of Texas M. D. Anderson Cancer Center, Department of Lymphoma & Myeloma, Houston, TX
- The University of Texas M. D. Anderson Cancer Center, Department of Experimental Therapeutics, Division of Cancer Medicine, Houston, TX
| |
Collapse
|
26
|
|
27
|
|