64 results on '"Orlowski, Robert"'
Search Results
2. Contributeurs
- Author
-
Runge, Marschall S., primary, Greganti, M. Andrew, additional, Adimora, Adaora A., additional, Alattar, Maha, additional, Aris, Robert M., additional, Bae-Jump, Victoria Lin, additional, Baggstrom, Maria Q., additional, Barritt, A. Sidney, additional, Bassim, Marc K., additional, Bates, Toby, additional, Beaven, Anne W., additional, Berger, Robert G., additional, Berkowitz, Lee R., additional, Bernard, Stephen A., additional, Blau, William S., additional, Boggess, John F., additional, Bowman, Mary C., additional, Brecher, Mark E., additional, Bromberg, Philip A., additional, Brown, Sue A., additional, Brown, Vickie, additional, Bryson, Paul C., additional, Buckmire, Robert A., additional, Bullitt, Elizabeth, additional, Burkhart, Craig, additional, Busby-Whitehead, M. Janette, additional, Buse, John B., additional, Bynum, Debra L., additional, Carey, Lisa A., additional, Carey, Timothy S., additional, Carson, Culley C., additional, Chang, Patricia P., additional, Chaudhary, Sanjay, additional, Clemmons, David R., additional, Coghill, James M., additional, Colindres, Romulo E., additional, Connolly, AnnaMarie, additional, Copeland, Benjamin J., additional, Correll, Todd, additional, Denu-Ciocca, Cynthia J., additional, Devetski, Thomas S., additional, DeWalt, Darren A., additional, Diaz, Luis A., additional, Donohue, James F., additional, Dooley, Mary Anne, additional, Dostou, Jean M., additional, Drossman, Douglas A., additional, Dupree, Carla Sueta, additional, Eapen, Rose J., additional, Ebert, Charles S., additional, Erdem, Nurum F., additional, Eron, Joseph J., additional, Falk, Ronald J., additional, Farmer-Boatwright, Mary Katherine, additional, Fasy, Elizabeth A., additional, Finkel, Alan G., additional, Finn, William F., additional, Fitzgerald, David P., additional, Ford, Carol A., additional, Forneris, Catherine A., additional, Fowler, Amy M., additional, Fowler, W. Craig, additional, Fowler, Wesley Caswell, additional, Fried, Michael W., additional, Gabriel, Don A., additional, Galvin, Shannon, additional, Gangarosa, Lisa M., additional, Garbutt, James C., additional, Gay, Cynthia, additional, Gaylord, Susan A., additional, Gettes, Leonard S., additional, Ghio, Andrew J., additional, Gilmore, John H., additional, Godley, Paul A., additional, Goldberg, Lee R., additional, Goldberg, Richard M., additional, Goldenberg, Matthew N., additional, Goldstein, Brian P., additional, Greenwood, Robert S., additional, Grimm, Ian S., additional, Grossman, Steven H., additional, Gwyther, Robert E., additional, Haggerty, John J., additional, Harris, Russell P., additional, Heizer, William D., additional, Henderson, Ashley G., additional, Henke, David C., additional, Hill, Michael A., additional, Hinderliter, Alan L., additional, Hinn, Albert R., additional, Hladik, Gerald A., additional, Hoffman, Hal M., additional, Hosseinipour, Mina C., additional, Howard, James F., additional, Huang, David Y., additional, Huang, Xuemei, additional, Hutto, Burton R., additional, Isaacs, Kim L., additional, Israel, Bruce F., additional, Ivester, Thomas S., additional, Jacobe, Heidi T., additional, Jacobson, Peter Lars, additional, Jantac, Lukas, additional, Jawanda, Jaspaul S., additional, Johnson, Sandra M., additional, Jonas, Beth L., additional, Jordan, Joanne M., additional, Juliano, Jonathan J., additional, Kahn, Kevin A., additional, Kaplan, Andrew H., additional, Key, Nigel S., additional, Kim, William Y., additional, Kizer, John S., additional, Klein, Caroline M., additional, Klemmer, Philip J., additional, Kölln, Karen, additional, Koruda, Mark J., additional, Kurz, James E., additional, LaCour, Jeffrey, additional, Ladha, Alim M., additional, Leight, W. Derek, additional, Leone, Peter A., additional, Lindsey, B. Anthony, additional, Madanick, Ryan D., additional, Mandelkehr, Lawrence K., additional, Mann, J. Douglas, additional, Markovic-Plese, Silva, additional, Marshall, Allen F., additional, Mattern, William D., additional, Mayer, Celeste M., additional, Meredith, Travis A., additional, Miller, William C., additional, Mitchell, Beverly S., additional, Moll, Stephan, additional, Morgan, Douglas R., additional, Morrell, Dean S., additional, Muñoz, M. Cristina, additional, Nachman, Patrick H., additional, Nelson, Kelly C., additional, Nester, Carla M., additional, Nicholas, Linda M., additional, Ohman, E. Magnus, additional, O'Neil, Bert H., additional, Ontjes, David A., additional, Orlowski, Robert Z., additional, Parsons, Daniel J., additional, Patel, Dhavalkumar D., additional, Patterson, Cam, additional, Patterson, Kristine B., additional, Peppercorn, Amanda, additional, Pillsbury, Harold C., additional, Rathmell, W. Kimryn, additional, Reuland, Daniel S., additional, Ringel, Yehuda, additional, Rivera, M. Patricia, additional, Rosebrock, Craig N., additional, Rosenberg, Pinchas, additional, Roubey, Robert A.S., additional, Rubenstein, David S., additional, Runge, Susan Riggs, additional, Russo, Mark, additional, Rutala, William A., additional, Sanders, William E., additional, Sanoff, Hanna K., additional, Sanoff, Scott L., additional, Scarlett, Yolanda V., additional, Schwarz, Emily J., additional, Senior, Brent A., additional, Serody, Jonathan S., additional, Shaheen, Nicholas J., additional, Shea, Thomas C., additional, Sheahan, Richard G., additional, Shockley, William W., additional, Shrestha, Roshan, additional, Sickbert-Bennett, Emily E., additional, Sickel, Micah J., additional, Sikich, Linmarie, additional, Simpson, Ross J., additional, Smith, Sidney C., additional, Socinski, Mark A., additional, Sparling, P. Frederick, additional, Stinchcombe, Thomas E., additional, Stouffer, George A., additional, Tarrant, Teresa K., additional, Taylor, Mark, additional, Thomas, Michael J., additional, Thomas, Nancy E., additional, Thorpe, John M., additional, Tilley, Stephen L., additional, Ting, Jenny P., additional, Tomsick, Robert S., additional, van der Horst, Charles M., additional, Vaughn, Bradley V., additional, Vick, Pamela G., additional, Vissers, Robert J., additional, Voorhees, Peter M., additional, Wang, Tracy Y., additional, Watson, Lea C., additional, Weber, David J., additional, Wehbie, Robert S., additional, Weissler, Mark C., additional, Wells, Ellen C., additional, Whang, Young E., additional, Willis, Park W., additional, Winfield, John B., additional, Winzelberg, Gary S., additional, Wohl, David A., additional, Wong, Leslie P., additional, Wu, Diem N., additional, and Zacks, Steven, additional
- Published
- 2011
- Full Text
- View/download PDF
3. Myélome multiple
- Author
-
Orlowski, Robert Z., primary and Gabriel, Don A., additional
- Published
- 2011
- Full Text
- View/download PDF
4. Contributors
- Author
-
Runge, Marschall S., primary, Greganti, M. Andrew, additional, Adimora, Adaora A., additional, Alattar, Maha, additional, Aris, Robert M., additional, Bae-Jump, Victoria Lin, additional, Baggstrom, Maria Q., additional, Barritt, A. Sidney, additional, Bassim, Marc K., additional, Bates, Toby, additional, Beaven, Anne W., additional, Berger, Robert G., additional, Berkowitz, Lee R., additional, Bernard, Stephen A., additional, Blau, William S., additional, Boggess, John F., additional, Bowman, Mary C., additional, Brecher, Mark E., additional, Bromberg, Philip A., additional, Brown, Sue A., additional, Brown, Vickie, additional, Bryson, Paul C., additional, Buckmire, Robert A., additional, Bullitt, Elizabeth, additional, Burkhart, Craig, additional, Busby-Whitehead, M. Janette, additional, Buse, John B., additional, Bynum, Debra L., additional, Carey, Lisa A., additional, Carey, Timothy S., additional, Carson, Culley C., additional, Chang, Patricia P., additional, Chaudhary, Sanjay, additional, Clemmons, David R., additional, Coghill, James M., additional, Colindres, Romulo E., additional, Connolly, AnnaMarie, additional, Copeland, Benjamin J., additional, Correll, Todd, additional, Denu-Ciocca, Cynthia J., additional, Devetski, Thomas S., additional, DeWalt, Darren A., additional, Diaz, Luis A., additional, Donohue, James F., additional, Dooley, Mary Anne, additional, Dostou, Jean M., additional, Drossman, Douglas A., additional, Dupree, Carla Sueta, additional, Eapen, Rose J., additional, Ebert, Charles S., additional, Erdem, Nurum F., additional, Eron, Joseph J., additional, Falk, Ronald J., additional, Farmer-Boatwright, Mary Katherine, additional, Fasy, Elizabeth A., additional, Finkel, Alan G., additional, Finn, William F., additional, Fitzgerald, David P., additional, Ford, Carol A., additional, Forneris, Catherine A., additional, Fowler, Amy M., additional, Fowler, W. Craig, additional, Fowler, Wesley Caswell, additional, Fried, Michael W., additional, Gabriel, Don A., additional, Galvin, Shannon, additional, Gangarosa, Lisa M., additional, Garbutt, James C., additional, Gay, Cynthia, additional, Gaylord, Susan A., additional, Gettes, Leonard S., additional, Ghio, Andrew J., additional, Gilmore, John H., additional, Godley, Paul A., additional, Goldberg, Lee R., additional, Goldberg, Richard M., additional, Goldenberg, Matthew N., additional, Goldstein, Brian P., additional, Greenwood, Robert S., additional, Grimm, Ian S., additional, Grossman, Steven H., additional, Gwyther, Robert E., additional, Haggerty, John J., additional, Harris, Russell P., additional, Heizer, William D., additional, Henderson, Ashley G., additional, Henke, David C., additional, Hill, Michael A., additional, Hinderliter, Alan L., additional, Hinn, Albert R., additional, Hladik, Gerald A., additional, Hoffman, Hal M., additional, Hosseinipour, Mina C., additional, Howard, James F., additional, Huang, David Y., additional, Huang, Xuemei, additional, Hutto, Burton R., additional, Isaacs, Kim L., additional, Israel, Bruce F., additional, Ivester, Thomas S., additional, Jacobe, Heidi T., additional, Jacobson, Peter Lars, additional, Jantac, Lukas, additional, Jawanda, Jaspaul S., additional, Johnson, Sandra M., additional, Jonas, Beth L., additional, Jordan, Joanne M., additional, Juliano, Jonathan J., additional, Kahn, Kevin A., additional, Kaplan, Andrew H., additional, Key, Nigel S., additional, Kim, William Y., additional, Kizer, John S., additional, Klein, Caroline M., additional, Klemmer, Philip J., additional, Kölln, Karen, additional, Koruda, Mark J., additional, Kurz, James E., additional, LaCour, Jeffrey, additional, Ladha, Alim M., additional, Leight, W. Derek, additional, Leone, Peter A., additional, Lindsey, B. Anthony, additional, Madanick, Ryan D., additional, Mandelkehr, Lawrence K., additional, Mann, J. Douglas, additional, Markovic-Plese, Silva, additional, Marshall, Allen F., additional, Mattern, William D., additional, Mayer, Celeste M., additional, Meredith, Travis A., additional, Miller, William C., additional, Mitchell, Beverly S., additional, Moll, Stephan, additional, Morgan, Douglas R., additional, Morrell, Dean S., additional, Muñoz, M. Cristina, additional, Nachman, Patrick H., additional, Nelson, Kelly C., additional, Nester, Carla M., additional, Nicholas, Linda M., additional, Ohman, E. Magnus, additional, O'Neil, Bert H., additional, Ontjes, David A., additional, Orlowski, Robert Z., additional, Parsons, Daniel J., additional, Patel, Dhavalkumar D., additional, Patterson, Cam, additional, Patterson, Kristine B., additional, Peppercorn, Amanda, additional, Pillsbury, Harold C., additional, Rathmell, W. Kimryn, additional, Reuland, Daniel S., additional, Ringel, Yehuda, additional, Rivera, M. Patricia, additional, Rosebrock, Craig N., additional, Rosenberg, Pinchas, additional, Roubey, Robert A.S., additional, Rubenstein, David S., additional, Runge, Susan Riggs, additional, Russo, Mark, additional, Rutala, William A., additional, Sanders, William E., additional, Sanoff, Hanna K., additional, Sanoff, Scott L., additional, Scarlett, Yolanda V., additional, Schwarz, Emily J., additional, Senior, Brent A., additional, Serody, Jonathan S., additional, Shaheen, Nicholas J., additional, Shea, Thomas C., additional, Sheahan, Richard G., additional, Shockley, William W., additional, Shrestha, Roshan, additional, Sickbert-Bennett, Emily E., additional, Sickel, Micah J., additional, Sikich, Linmarie, additional, Simpson, Ross J., additional, Smith, Sidney C., additional, Socinski, Mark A., additional, Sparling, P. Frederick, additional, Stinchcombe, Thomas E., additional, Stouffer, George A., additional, Tarrant, Teresa K., additional, Taylor, Mark, additional, Thomas, Michael J., additional, Thomas, Nancy E., additional, Thorpe, John M., additional, Tilley, Stephen L., additional, Ting, Jenny P., additional, Tomsick, Robert S., additional, van der Horst, Charles M., additional, Vaughn, Bradley V., additional, Vick, Pamela G., additional, Vissers, Robert J., additional, Voorhees, Peter M., additional, Wang, Tracy Y., additional, Watson, Lea C., additional, Weber, David J., additional, Wehbie, Robert S., additional, Weissler, Mark C., additional, Wells, Ellen C., additional, Whang, Young E., additional, Willis, Park W., additional, Winfield, John B., additional, Winzelberg, Gary S., additional, Wohl, David A., additional, Wong, Leslie P., additional, Wu, Diem N., additional, and Zacks, Steven, additional
- Published
- 2009
- Full Text
- View/download PDF
5. Malignant Lymphomas
- Author
-
Voorhees, Peter M., primary and Orlowski, Robert Z., additional
- Published
- 2009
- Full Text
- View/download PDF
6. Multiple Myeloma
- Author
-
Orlowski, Robert Z., primary and Gabriel, Don A., additional
- Published
- 2009
- Full Text
- View/download PDF
7. Resensitising proteasome inhibitor-resistant myeloma with sphingosine kinase 2 inhibition
- Author
-
Briony L. Gliddon, Craig T. Wallington-Beddoe, Manjun Li, Darren J. Creek, Melissa R. Pitman, Melinda N. Tea, Paul Wang, Dovile Anderson, John Toubia, Melissa K. Bennett, Robert Z. Orlowski, Stuart M. Pitson, Jason A. Powell, Bennett, Melissa K, Li, Manjun, Tea, Melinda N, Pitman, Melissa R, Toubia, John, Wang, Paul PS, Anderson, Dovile, Creek, Darren J, Orlowski, Robert Z, Gliddon, Briony L, Powell, Jason A, Wallington-Beddoe, Craig T, and Pitson, Stuart M
- Subjects
Cancer Research ,ATF4, activating transcription factor 4 ,Resistance ,Myeloma ,medicine.disease_cause ,Sphingolipid ,Bortezomib ,Unfolded protein response ,Gene Knockout Techniques ,Mice ,chemistry.chemical_compound ,UPR, unfolded protein response ,GSEA, gene set enrichment analysis ,immune system diseases ,hemic and lymphatic diseases ,Enzyme Inhibitors ,RC254-282 ,Mutation ,bortezomib ,Sphingosine Kinase 2 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,unfolded protein response ,BR, bortezomib resistant ,Phosphotransferases (Alcohol Group Acceptor) ,myeloma ,S1P, sphingosine 1-phosphate ,Multiple Myeloma ,Proteasome Inhibitors ,medicine.drug ,wt, wild-type ,Original article ,ATF6, activating transcription factor 6 ,Cell Survival ,IRE1, inositol-requiring enzyme 1 ,Antineoplastic Agents ,resistance ,ER, endoplasmic reticulum ,Structure-Activity Relationship ,Cell Line, Tumor ,medicine ,Animals ,Humans ,cardiovascular diseases ,neoplasms ,SK2, sphingosine kinase 2 ,Dose-Response Relationship, Drug ,business.industry ,PERK, protein kinase R-like ER kinase ,XBP1s, X-box binding protein 1s ,Xenograft Model Antitumor Assays ,Carfilzomib ,CR, carfilzomib resistant ,Disease Models, Animal ,Proteasome ,chemistry ,Drug Resistance, Neoplasm ,Proteasome inhibitor ,Cancer research ,sphingolipid ,business - Abstract
Refereed/Peer-reviewed The introduction of the proteasome inhibitor bortezomib into treatment regimens for myeloma has led to substantial improvement in patient survival. However, whilst bortezomib elicits initial responses in many myeloma patients, this haematological malignancy remains incurable due to the development of acquired bortezomib resistance. With other patients presenting with disease that is intrinsically bortezomib resistant, it is clear that new therapeutic approaches are desperately required to target bortezomib-resistant myeloma. We have previously shown that targeting sphingolipid metabolism with the sphingosine kinase 2 (SK2) inhibitor K145 in combination with bortezomib induces synergistic death of bortezomib-naive myeloma. In the current study, we have demonstrated that targeting sphingolipid metabolism with K145 synergises with bortezomib and effectively resensitises bortezomib-resistant myeloma to this proteasome inhibitor Notably, these effects were dependent on enhanced activation of the unfolded protein response, and were observed in numerous separate myeloma models that appear to have different mechanisms of bortezomib resistance, including a new bortezomib-resistant myeloma model we describe which possesses a clinically relevant proteasome mutation. Furthermore, K145 also displayed synergy with the next-generation proteasome inhibitor carfilzomib in bortezomib-resistant and carfilzomib-resistant myeloma cells. Together, these findings indicate that targeting sphingolipid metabolism via SK2 inhibition may be effective in combination with a broad spectrum of proteasome inhibitors in the proteasome inhibitor resistant setting, and is an approach worth clinical exploration.
- Published
- 2022
8. Dexamethasone dose intensity does not impact outcomes in newly diagnosed multiple myeloma: a secondary SWOG analysis.
- Author
-
Banerjee R, Sexton R, Cowan AJ, Rosenberg AS, Ailawadhi S, Rajkumar SV, Kumar SK, Dispenzieri A, Lonial S, Durie BGM, Richardson PG, Usmani SZ, Hoering A, and Orlowski RZ
- Abstract
Dexamethasone is a key component of induction for newly diagnosed multiple myeloma (NDMM) despite common toxicities including hyperglycemia and insomnia. In the randomized ECOG E4A03 trial, dexamethasone 40 milligrams (mg) once weekly was associated with lower mortality than higher doses of dexamethasone. However, the performance of dexamethasone dose reductions below this threshold with regard to progression-free survival (PFS) and overall survival (OS) in NDMM have not been fully characterized. We conducted a secondary pooled analysis of the S0777 and S1211 SWOG studies of NDMM, which employed lenalidomide-dexamethasone (Rd) alone with or without bortezomib (VRd) and with or without elotuzumab (Elo-VRd). Planned dexamethasone intensity was 40-60 mg weekly in all arms. Patients were categorized into FD-DEX (full-dose dexamethasone maintained throughout induction) or LD-DEX (lowered-dose dexamethasone or discontinuation; only permitted for Grade 3+ toxicities per both study protocols). Of 541 evaluated patients, the LD-DEX group comprised 373 patients (69%). There was no difference in PFS or OS between the FD-DEX or LD-DEX groups, which were balanced in terms of age, stage, and performance status. Predictors of PFS and OS in multivariate models were treatment arm, age ≥70, and thrombocytopenia; FD-DEX did not significantly improve either outcome. Our study suggests that dexamethasone dose reductions are common in multiple myeloma, even within clinical trials. Given dexamethasone's many toxicities and unclear benefit in the era of modern treatment regimens, dexamethasone dose reduction during NDMM induction warrants further prospective study. NCT00644228, NCT01668719., (Copyright © 2024 American Society of Hematology.)
- Published
- 2024
- Full Text
- View/download PDF
9. A randomized phase 2 trial of idiotype vaccination and adoptive autologous T-cell transfer in patients with multiple myeloma.
- Author
-
Qazilbash MH, Saini NY, Cha SC, Wang Z, Stadtmauer EA, Baladandayuthapani V, Lin H, Tross B, Honhar M, Rao SS, Kim K, Popescu M, Szymura S, Zhang T, Anderson A, Bashir Q, Shpall EJ, Orlowski RZ, Levine BL, Kerr N, Garfall A, Cohen A, Vogl DT, Dengel K, June CH, Champlin R, and Kwak LW
- Subjects
- Autografts, Cancer Vaccines immunology, Disease-Free Survival, Female, Hemocyanins administration & dosage, Hemocyanins immunology, Humans, Male, Survival Rate, Transplantation, Autologous, Adoptive Transfer, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes transplantation, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes transplantation, Cancer Vaccines administration & dosage, Memory T Cells immunology, Memory T Cells transplantation, Multiple Myeloma immunology, Multiple Myeloma mortality, Multiple Myeloma therapy, Vaccination
- Abstract
We hypothesized that combining adoptively transferred autologous T cells with a cancer vaccine strategy would enhance therapeutic efficacy by adding antimyeloma idiotype (Id)-keyhole limpet hemocyanin (KLH) vaccine to vaccine-specific costimulated T cells. In this randomized phase 2 trial, patients received either control (KLH only) or Id-KLH vaccine, autologous transplantation, vaccine-specific costimulated T cells expanded ex vivo, and 2 booster doses of assigned vaccine. In 36 patients (KLH, n = 20; Id-KLH, n = 16), no dose-limiting toxicity was seen. At last evaluation, 6 (30%) and 8 patients (50%) had achieved complete remission in KLH-only and Id-KLH arms, respectively (P = .22), and no difference in 3-year progression-free survival was observed (59% and 56%, respectively; P = .32). In a 594 Nanostring nCounter gene panel analyzed for immune reconstitution (IR), compared with patients receiving KLH only, there was a greater change in IR genes in T cells in those receiving Id-KLH relative to baseline. Specifically, upregulation of genes associated with activation, effector function induction, and memory CD8+ T-cell generation after Id-KLH but not after KLH control vaccination was observed. Similarly, in responding patients across both arms, upregulation of genes associated with T-cell activation was seen. At baseline, all patients had greater expression of CD8+ T-cell exhaustion markers. These changes were associated with functional Id-specific immune responses in a subset of patients receiving Id-KLH. In conclusion, in this combination immunotherapy approach, we observed significantly more robust IR in CD4+ and CD8+ T cells in the Id-KLH arm, supporting further investigation of vaccine and adoptive immunotherapy strategies. This trial was registered at www.clinicaltrials.gov as #NCT01426828., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
10. Experience with denosumab (XGEVA®) for prevention of skeletal-related events in the 10 years after approval.
- Author
-
Cadieux B, Coleman R, Jafarinasabian P, Lipton A, Orlowski RZ, Saad F, Scagliotti GV, Shimizu K, and Stopeck A
- Abstract
Skeletal-related events (SREs) are complications of bone metastases and carry a significant patient and economic burden. Denosumab is a receptor activator of nuclear factor-κB ligand (RANKL) inhibitor approved for SRE prevention in patients with multiple myeloma and patients with bone metastases from solid tumors. In phase 3 trials, denosumab showed superiority to the bisphosphonate zoledronate in reducing the risk of first on-study SRE by 17% (median time to first on-study SRE delayed by 8.2 months) and the risk of first and subsequent on-study SREs by 18% across multiple solid tumor types, including some patients with multiple myeloma. Denosumab also improved pain outcomes and reduced the need for strong opioids. Additionally, a phase 3 trial showed denosumab was noninferior to zoledronate in delaying time to first SRE in patients with newly diagnosed multiple myeloma. Denosumab has a convenient 120 mg every 4 weeks recommended dosing schedule with subcutaneous administration. Rare but serious toxicities associated with denosumab include osteonecrosis of the jaw, hypocalcemia, and atypical femoral fracture events, with multiple vertebral fractures reported following treatment discontinuation. After a decade of real-world clinical experience with denosumab, we are still learning about the optimal use and dosing for denosumab. Despite the emergence of novel and effective antitumor therapies, there remains a strong rationale for the clinical utility of antiresorptive therapy for SRE prevention. Ongoing studies aim to optimize clinical management of patients using denosumab for SRE prevention while maintaining safety and efficacy., Competing Interests: The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Benoit Cadieux was an employee and shareholder of Amgen Inc. at the time of the development of this review. Robert Coleman has received lecture fees from Amgen and Novartis; consultancy fees from Amgen, Astellas, AstraZeneca, Boehringer Ingelheim, ITM, and Scancell. Pegah Jafarinasabian is an employee and shareholder of Amgen Inc. Allan Lipton has no financial interests or personal relationships that may be considered as potential competing interests. Robert Z. Orlowski has no financial interests or personal relationships that may be considered as potential competing interests.. Fred Saad served as a consultant, advisory board member and received honoraria and research funding from Amgen; he has also served as a consultant, advisory board member and received honoraria and research funding from Astellas, AstraZeneca, Bayer, Janssen, Myovant, Novartis, Pfizer, and Sanofi. Giorgio V. Scagliotti has no financial interests or personal relationships that may be considered as potential competing interests. Kazuyuki Shimiz has no financial interests or personal relationships that may be considered as potential competing interests. Alison Stopeck has received consulting fees from Amgen and AstraZeneca; contracted research support from Amgen, Exact Sciences; speakers bureau from Exact Sciences; and honoraria from Amgen., (© 2022 The Authors.)
- Published
- 2022
- Full Text
- View/download PDF
11. Resensitising proteasome inhibitor-resistant myeloma with sphingosine kinase 2 inhibition.
- Author
-
Bennett MK, Li M, Tea MN, Pitman MR, Toubia J, Wang PP, Anderson D, Creek DJ, Orlowski RZ, Gliddon BL, Powell JA, Wallington-Beddoe CT, and Pitson SM
- Subjects
- Animals, Antineoplastic Agents chemistry, Antineoplastic Agents therapeutic use, Bortezomib chemistry, Bortezomib pharmacology, Cell Line, Tumor, Cell Survival drug effects, Disease Models, Animal, Dose-Response Relationship, Drug, Enzyme Inhibitors therapeutic use, Gene Knockout Techniques, Humans, Mice, Multiple Myeloma drug therapy, Multiple Myeloma genetics, Multiple Myeloma metabolism, Multiple Myeloma pathology, Proteasome Inhibitors chemistry, Proteasome Inhibitors therapeutic use, Structure-Activity Relationship, Unfolded Protein Response drug effects, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Drug Resistance, Neoplasm drug effects, Enzyme Inhibitors pharmacology, Phosphotransferases (Alcohol Group Acceptor) antagonists & inhibitors, Proteasome Inhibitors pharmacology
- Abstract
The introduction of the proteasome inhibitor bortezomib into treatment regimens for myeloma has led to substantial improvement in patient survival. However, whilst bortezomib elicits initial responses in many myeloma patients, this haematological malignancy remains incurable due to the development of acquired bortezomib resistance. With other patients presenting with disease that is intrinsically bortezomib resistant, it is clear that new therapeutic approaches are desperately required to target bortezomib-resistant myeloma. We have previously shown that targeting sphingolipid metabolism with the sphingosine kinase 2 (SK2) inhibitor K145 in combination with bortezomib induces synergistic death of bortezomib-naïve myeloma. In the current study, we have demonstrated that targeting sphingolipid metabolism with K145 synergises with bortezomib and effectively resensitises bortezomib-resistant myeloma to this proteasome inhibitor. Notably, these effects were dependent on enhanced activation of the unfolded protein response, and were observed in numerous separate myeloma models that appear to have different mechanisms of bortezomib resistance, including a new bortezomib-resistant myeloma model we describe which possesses a clinically relevant proteasome mutation. Furthermore, K145 also displayed synergy with the next-generation proteasome inhibitor carfilzomib in bortezomib-resistant and carfilzomib-resistant myeloma cells. Together, these findings indicate that targeting sphingolipid metabolism via SK2 inhibition may be effective in combination with a broad spectrum of proteasome inhibitors in the proteasome inhibitor resistant setting, and is an approach worth clinical exploration., Competing Interests: Declaration of competing interests The authors declare that they have no competing interests., (Copyright © 2021. Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
12. Genetic determinants of multiple myeloma risk within the Wnt/beta-catenin signaling pathway.
- Author
-
Belachew AA, Wu X, Callender R, Waller R, Orlowski RZ, Vachon CM, Camp NJ, Ziv E, and Hildebrandt MAT
- Subjects
- Black or African American genetics, Black or African American statistics & numerical data, Aged, Case-Control Studies, Female, Genetic Predisposition to Disease ethnology, Hispanic or Latino genetics, Hispanic or Latino statistics & numerical data, Humans, Male, Middle Aged, White People genetics, White People statistics & numerical data, Multiple Myeloma ethnology, Multiple Myeloma genetics, Wnt Signaling Pathway genetics, beta Catenin genetics
- Abstract
Background: Aberrant Wnt/beta-catenin pathway activation is implicated in Multiple Myeloma (MM) development, but little is known if genetic variants within this pathway contribute to MM susceptibility., Methods: We performed a discovery candidate pathway analysis in 269 non-Hispanic white MM cases and 272 controls focusing on 171 variants selected from 26 core genes within the Wnt/beta-catenin pathway. Significant candidate variants (P < 0.05) were selected for validation in internal and external non-Hispanic white populations totaling 818 cases and 1209 controls. We also examined significant variants in non-Hispanic black and Hispanic case/control study populations to identify potential differences by race/ethnicity. Possible biological functions of candidate variants were predicted in silico., Results: Seven variants were significantly associated with MM risk in non-Hispanic whites in the discovery population, of which LRP6:rs7966410 (OR: 0.57; 95 % CI: 0.38-0.88; P = 9.90 × 10
-3 ) and LRP6:rs7956971 (OR: 0.64; 95 % CI: 0.44-0.95; P = 0.027) remained significant in the internal and external populations. CSNK1D:rs9901910 replicated among all three racial/ethnic groups, with 2-6 fold increased risk of MM (OR: 2.40; 95 % CI: 1.67-3.45; P = 2.43 × 10-6 - non-Hispanic white; OR: 6.42; 95 % CI: 2.47-16.7; P = 3.14 × 10-4 - non-Hispanic black; OR: 4.31; 95 % CI: 1.83-10.1; P = 8.10 × 10-4 - Hispanic). BTRC:rs7916830 was associated with a significant 37 % and 24 % reduced risk of MM in the non-Hispanic white (95 % CI: 0.49-0.82; P = 5.60 × 10-4 ) and non-Hispanic Black (95 % CI: 0.60-0.97; P = 0.028) population, respectively. In silico tools predicted that these loci altered function through via gene regulation., Conclusion: We identified several variants within the Wnt/beta-catenin pathway associated with MM susceptibility. Findings of this study highlight the potential genetic role of Wnt/beta-catenin signaling in MM etiology among a diverse patient population., (Copyright © 2021 Elsevier Ltd. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
13. Molecular and genetic biomarkers implemented from next-generation sequencing provide treatment insights in clinical practice for Waldenström macroglobulinemia.
- Author
-
Wang Y, Gali VL, Xu-Monette ZY, Sano D, Thomas SK, Weber DM, Zhu F, Fang X, Deng M, Zhang M, Hagemeister FB, Li Y, Orlowski RZ, Lee HC, and Young KH
- Subjects
- Adenine analogs & derivatives, Adenine therapeutic use, Adult, Agammaglobulinaemia Tyrosine Kinase antagonists & inhibitors, Aged, Aged, 80 and over, DNA Mutational Analysis, Dexamethasone therapeutic use, Female, Genetic Testing, High-Throughput Nucleotide Sequencing, Humans, Immunoglobulin A metabolism, Immunoglobulin G metabolism, Immunoglobulin M metabolism, Male, Middle Aged, Piperidines therapeutic use, Polymerase Chain Reaction, Prognosis, Rituximab therapeutic use, Sequence Analysis, DNA, Antineoplastic Agents therapeutic use, Myeloid Differentiation Factor 88 genetics, Receptors, CXCR4 genetics, Tumor Suppressor Protein p53 genetics, Waldenstrom Macroglobulinemia drug therapy, Waldenstrom Macroglobulinemia genetics
- Abstract
Waldenström macroglobulinemia (WM) is a distinct type of indolent lymphoplasmacytic lymphoma (LPL) with a high frequency of MYD88
L265P mutation. Treatment for WM/LPL is highly variable in clinic and ibrutinib (a Bruton tyrosine kinase inhibitor, BTKi) has become a new treatment option for WM. To investigate the clinical impact of genetic alterations in WM, we assembled a large cohort of 219 WMs and 12 LPLs dividing into two subcohorts: a training cohort, patients sequenced by a same targeted 29-gene next-generation sequencing (NGS) panel, and a validation cohort, patients sequenced by allele specific-PCR or other targeted NGS panels. In both training and validation subcohorts, MYD88L265P and TP53 mutations showed favorable and adverse prognostic effects, respectively. CXCR4 nonsense/missense mutations (CXCR4NS/MS ), cytogenetic complex karyotypes, and a family history of lymphoma/leukemia in first-degree relatives were associated with significantly worse clinical outcomes only or more in the validation subcohort. We further investigated the efficacy of various treatments and interaction with genetic factors in the entire cohort. Upfront dexamethasone usage was associated with poorer clinical outcomes in patients who received non-proteasome-containing chemotherapy as first-line treatment independent of genetic factors. Maintenance rituximab was associated with better survival. Ibrutinib/BTKi showed potential benefit in relapsed/refractory patients and patients without CXCR4NS/MS including those with TP53 mutations. In conclusion, genetic testing for MYD88L265P , TP53, and CXCR4 mutations and cytogenetic analysis provide important information for prognosis prediction and therapy selection. The findings in these study are valuable for improving treatment decisions on therapies available for WM/LPL patients with integration of NGS in clinic., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
14. Changes in Field Termination of Resuscitation and Survival Rates After an Educational Intervention to Promote on Scene Resuscitation for Out-of-Hospital Cardiac Arrest.
- Author
-
Clemency BM, Innes JC, Waldrop M, White LJ, Dievendorf E, Orlowski R, Wang K, Lindstrom HA, Canty JM Jr, and Hostler D
- Subjects
- Humans, Retrospective Studies, Survival Rate, Cardiopulmonary Resuscitation, Emergency Medical Services, Out-of-Hospital Cardiac Arrest therapy
- Abstract
Background: Emergency medical services (EMS) agencies with higher field termination-of-resuscitation (TOR) rates tend to have higher survival rates from out-of-hospital cardiac arrest (OHCA). Whether EMS agencies can improve survival rates through efforts to focus on resuscitation on scene and optimize TOR rates is unknown., Objective: The goal of this study was to determine if an EMS agency's efforts to enhance on-scene resuscitation were associated with increased TOR and OHCA survival with favorable neurologic outcome., Methods: A single-city, retrospective analysis of prospectively collected 2017 quality assurance data was conducted. Patient demographics, process, and outcome measures were compared before and after an educational intervention to increase field TOR. The primary outcome measure was survival to hospital discharge with favorable neurologic status., Results: There were 320 cases that met inclusion criteria. No differences in age, gender, location, witnessed arrest, bystander cardiopulmonary resuscitation, initial shockable rhythm, or presumed cardiac etiology were found. After the intervention, overall TOR rate increased from 39.6% to 51.1% (p = 0.06). Among subjects transported without return of spontaneous circulation (ROSC), average time on scene increased from 26.4 to 34.2 min (p = 0.02). Rates of sustained ROSC and survival to hospital admission were similar between periods. After intervention, there was a trend toward increased survival to hospital discharge rate (relative risk [RR] 2.09; 95% confidence interval [CI] 0.74-5.91) and an increase in survival with favorable neurologic status rate (RR 5.96; 95% CI 0.80-44.47)., Conclusion: This study described the association between an educational intervention focusing on optimization of resuscitation on scene and OHCA process and outcome measures. Field termination has the potential to serve as a surrogate marker for aggressively treating OHCA patients on scene., (Copyright © 2020. Published by Elsevier Inc.)
- Published
- 2021
- Full Text
- View/download PDF
15. Phase 1 Trial Evaluating Vorinostat Plus Bortezomib, Lenalidomide, and Dexamethasone in Patients With Newly Diagnosed Multiple Myeloma.
- Author
-
Kaufman JL, Mina R, Shah JJ, Laubach JP, Nooka AK, Lewis C, Gleason C, Sharp C, Harvey RD, Heffner LT, Richardson P, Lonial S, and Orlowski RZ
- Subjects
- Adult, Aged, Bortezomib pharmacology, Dexamethasone pharmacology, Female, Humans, Lenalidomide pharmacology, Male, Middle Aged, Multiple Myeloma pathology, Vorinostat pharmacology, Bortezomib therapeutic use, Dexamethasone therapeutic use, Lenalidomide therapeutic use, Multiple Myeloma drug therapy, Vorinostat therapeutic use
- Abstract
Introduction: Bortezomib plus lenalidomide and dexamethasone (VRD) is a standard induction therapy for newly diagnosed multiple myeloma (NDMM) patients. Given preclinical and clinical data suggesting the synergistic activity of the histone deacetylase inhibitor vorinostat with both bortezomib and lenalidomide for the treatment of multiple myeloma, we hypothesized that adding vorinostat to VRD (R2V2) would increase the rate and the quality of responses to induction treatment. Here we report the results of a phase 1 trial (NCT01038388) evaluating R2V2 as up-front treatment for NDMM patients., Patients and Methods: R2V2 was tested as induction therapy in a dose-escalation phase 1 study in 30 NDMM patients deemed eligible for autologous stem-cell transplantation. Treatment consisted of 4 induction cycles with R2V2, followed by either autologous stem-cell transplantation or 4 additional R2V2 cycles and lenalidomide maintenance therapy., Results: The maximum tolerated dose of vorinostat was 200 mg daily. The most common adverse events were gastrointestinal (87%), fatigue and peripheral neuropathy (60%), and thrombocytopenia (33%). R2V2 induced an objective response in 96% of patients, with 48% obtaining at least a complete remission. Median progression-free survival was 52 months, with 77% of patients alive at 5 years., Conclusion: R2V2 as induction treatment for NDMM patients resulted in remarkable response rates at the cost of increased toxicity., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
16. Daratumumab, lenalidomide, bortezomib, and dexamethasone for transplant-eligible newly diagnosed multiple myeloma: the GRIFFIN trial.
- Author
-
Voorhees PM, Kaufman JL, Laubach J, Sborov DW, Reeves B, Rodriguez C, Chari A, Silbermann R, Costa LJ, Anderson LD Jr, Nathwani N, Shah N, Efebera YA, Holstein SA, Costello C, Jakubowiak A, Wildes TM, Orlowski RZ, Shain KH, Cowan AJ, Murphy S, Lutska Y, Pei H, Ukropec J, Vermeulen J, de Boer C, Hoehn D, Lin TS, and Richardson PG
- Subjects
- Adult, Aged, Antibodies, Monoclonal adverse effects, Bortezomib adverse effects, Combined Modality Therapy, Female, Humans, Lenalidomide adverse effects, Maintenance Chemotherapy methods, Male, Middle Aged, Multiple Myeloma drug therapy, Multiple Myeloma mortality, Multiple Myeloma pathology, Patient Selection, Transplantation, Autologous, Antibodies, Monoclonal administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bortezomib administration & dosage, Dexamethasone administration & dosage, Hematopoietic Stem Cell Transplantation, Lenalidomide administration & dosage, Multiple Myeloma therapy
- Abstract
Lenalidomide, bortezomib, and dexamethasone (RVd) followed by autologous stem cell transplantation (ASCT) is standard frontline therapy for transplant-eligible patients with newly diagnosed multiple myeloma (NDMM). The addition of daratumumab (D) to RVd (D-RVd) in transplant-eligible NDMM patients was evaluated. Patients (N = 207) were randomized 1:1 to D-RVd or RVd induction (4 cycles), ASCT, D-RVd or RVd consolidation (2 cycles), and lenalidomide or lenalidomide plus D maintenance (26 cycles). The primary end point, stringent complete response (sCR) rate by the end of post-ASCT consolidation, favored D-RVd vs RVd (42.4% vs 32.0%; odds ratio, 1.57; 95% confidence interval, 0.87-2.82; 1-sided P = .068) and met the prespecified 1-sided α of 0.10. With longer follow-up (median, 22.1 months), responses deepened; sCR rates improved for D-RVd vs RVd (62.6% vs 45.4%; P = .0177), as did minimal residual disease (MRD) negativity (10-5 threshold) rates in the intent-to-treat population (51.0% vs 20.4%; P < .0001). Four patients (3.8%) in the D-RVd group and 7 patients (6.8%) in the RVd group progressed; respective 24-month progression-free survival rates were 95.8% and 89.8%. Grade 3/4 hematologic adverse events were more common with D-RVd. More infections occurred with D-RVd, but grade 3/4 infection rates were similar. Median CD34+ cell yield was 8.2 × 106/kg for D-RVd and 9.4 × 106/kg for RVd, although plerixafor use was more common with D-RVd. Median times to neutrophil and platelet engraftment were comparable. Daratumumab with RVd induction and consolidation improved depth of response in patients with transplant-eligible NDMM, with no new safety concerns. This trial was registered at www.clinicaltrials.gov as #NCT02874742., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
17. Minimal Residual Disease Negativity Does Not Overcome Poor Prognosis in High-Risk Multiple Myeloma: A Single-Center Retrospective Study.
- Author
-
Kunacheewa C, Lee HC, Patel K, Thomas S, Amini B, Srour S, Bashir Q, Nieto Y, Qazilbash MH, Weber DM, Feng L, Orlowski RZ, Lin P, and Manasanch EE
- Subjects
- Adult, Aged, Aged, 80 and over, Disease-Free Survival, Female, Humans, Male, Middle Aged, Neoplasm, Residual, Retrospective Studies, Survival Rate, Multiple Myeloma blood, Multiple Myeloma mortality, Multiple Myeloma therapy
- Abstract
Background: Minimal residual disease (MRD) is a standard measurement for response assessment in multiple myeloma (MM). Despite new treatments, high-risk MM patients continue to have poor prognosis. We evaluated the effect of MRD negativity in high-risk versus standard-risk patients., Patients and Methods: We retrospectively evaluated all consecutive MM patients who underwent routine MRD testing by 1-tube 8-color advanced flow cytometry with 2,000,000 events and sensitivity level 10
-5 at our center from 2015 to 2018 after initial therapy. Kaplan-Meier and log-rank test were used to assess survival estimates and differences between study groups., Results: One hundred thirty-six patients with MRD testing after initial therapy or autologous stem-cell transplantation were identified. At a median follow-up of 14 months (range, 1-36 months), progression-free survival and overall survival were significantly worse in high-risk versus standard-risk patients. During the study period, 50% of high-risk group had experienced disease progression (relapse and/or death) versus 20% in the standard-risk group (P = .0006). No patients with standard-risk died, but 4 (14%) in the high-risk group did (P = .0007). Regardless of MRD status, high-risk patients had statistically significant worse progression-free survival than standard-risk patients. At median follow-up, those with disease 10% standard-risk/MRD negative; 20% standard-risk/MRD positive; 40% high-risk/MRD negative; and 45% high-risk/MRD positive had either experienced relapse or died (P = .0041). MRD status did not significantly affect overall survival in either group (P = .0914); however, longer follow-up is needed to assess survival., Conclusion: Genetic abnormalities remain a powerful prognostic indicator for MM, regardless of MRD status. For newly diagnosed MM patients treated with novel triple-drug initial therapy and frontline autologous stem-cell transplantation, MRD-negative status did not mitigate the poor-prognosis outcomes of high-risk MM patients., (Copyright © 2020 Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
18. Retrospective Review of the Use of High-Dose Cyclophosphamide, Bortezomib, Doxorubicin, and Dexamethasone for the Treatment of Multiple Myeloma and Plasma Cell Leukemia.
- Author
-
Tabchi S, Nair R, Kunacheewa C, Patel KK, Lee HC, Thomas SK, Amini B, Ahmed S, Mehta RS, Bashir Q, Qazilbash MH, Weber DM, Orlowski RZ, Alexanian R, Feng L, and Manasanch EE
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Bortezomib administration & dosage, Cyclophosphamide administration & dosage, Dexamethasone administration & dosage, Doxorubicin administration & dosage, Drug Resistance, Neoplasm, Female, Humans, Leukemia, Plasma Cell diagnosis, Leukemia, Plasma Cell mortality, Male, Middle Aged, Multiple Myeloma diagnosis, Multiple Myeloma mortality, Neoplasm Staging, Prognosis, Recurrence, Research Design, Retreatment, Treatment Outcome, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Leukemia, Plasma Cell drug therapy, Multiple Myeloma drug therapy
- Abstract
Background: Multiple myeloma (MM) usually follows a clinical course leading to refractoriness and limited treatment options in advanced stages, which might need bridge therapies to either autologous stem cell transplantation or novel therapies. We report our experience with the high-dose chemotherapy mCBAD (modified cyclophosphamide, bortezomib, doxorubicin, and dexamethasone) regimen in newly diagnosed MM (NDMM), relapsed/refractory MM (RRMM), and plasma cell leukemia (PCL) patients., Patients and Methods: We searched our electronic records database for MM patients who received mCBAD from 2010 to 2016 for 28-day cycles of cyclophosphamide 350 mg/m
2 intravenously (I.V.) twice daily with mesna 400 mg/m2 I.V. daily (days 1-4), bortezomib 1.3 mg/m2 subcutaneously/I.V. (days 1, 4, 8, 11), doxorubicin 9 mg/m2 daily continuous infusion (days 1-4), dexamethasone 40 mg orally daily (on days 1-4, 9-12, 17-20). International Myeloma Working Group (IMWG) criteria were used for response assessment and diagnosis. Descriptive statistics, Fisher exact test, χ2 , Wilcoxon rank sum, and Kaplan-Meier were used for statistical purposes., Results: One hundred forty patients met the inclusion criteria. A median of 2 cycles of therapy was administered. The overall response rate was 85% in patients with RRMM (n = 116) and 100% in NDMM (n = 13) and PCL (n = 11) patients. Respective median progression-free survival (mPFS) for NDMM, PCL, and RRMM were 19.61 months (95% confidence interval [CI], 5.26 to not applicable [NA]), 7.56 months (95% CI, 4.7 to NA), and 4.64 months (95% CI, 3.75-6.73). Patients with RRMM who used mCBAD as a bridge to autologous transplant (36.2%) had mPFS (11.48 months; 95% CI, 7.52-15.9 months) compared with those who did not (mPFS: 3.19 months; 95% CI, 2.4-3.75 months). Cytopenias occurred in more than 90% of patients, and febrile neutropenia was noted in 26%. All cases of treatment-related mortality (8%) occurred in patients with RRMM, except for 1 patient with PCL., Conclusion: mCBAD results in high response rates in myeloma and PCL, however, with high treatment-related mortality. Its use in RRMM should be limited to patients who have immediate need for therapy without other treatment options and who have good performance status (score of 0-1) or NDMM if novel agents are not available depending on practice setting. mCBAD can be a treatment option for patients with PCL., (Copyright © 2019 Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
19. Carfilzomib-Dexamethasone Versus Bortezomib-Dexamethasone in Relapsed or Refractory Multiple Myeloma: Updated Overall Survival, Safety, and Subgroups.
- Author
-
Orlowski RZ, Moreau P, Niesvizky R, Ludwig H, Oriol A, Chng WJ, Goldschmidt H, Yang Z, Kimball AS, and Dimopoulos M
- Subjects
- Aged, Bortezomib administration & dosage, Dexamethasone administration & dosage, Female, Follow-Up Studies, Humans, Male, Multiple Myeloma drug therapy, Multiple Myeloma pathology, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local pathology, Oligopeptides administration & dosage, Patient Safety, Prognosis, Survival Rate, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Drug Resistance, Neoplasm drug effects, Multiple Myeloma mortality, Neoplasm Recurrence, Local mortality, Salvage Therapy
- Abstract
Introduction: The phase III RandomizEd, OpeN Label, Phase 3 Study of Carfilzomib Plus DExamethAsone Vs Bortezomib Plus DexamethasOne in Patients With Relapsed Multiple Myeloma (ENDEAVOR) trial showed significantly improved progression-free survival and overall survival (OS) with carfilzomib (56 mg/m
2 ) and dexamethasone (Kd56) versus bortezomib and Kd56 (Vd) in patients with relapsed or refractory multiple myeloma (RRMM). We report updated OS and safety data after 6 months of additional follow-up., Patients and Methods: Patients with RRMM (1-3 previous lines of therapy) were randomized 1:1 to Kd56 or Vd. Median OS was estimated using the Kaplan-Meier method; OS was compared between treatment groups using Cox proportional hazards models., Results: As of July 19, 2017, median follow-up was 44.3 months for Kd56 and 43.7 months for Vd. Median OS was 47.8 months (Kd56) versus 38.8 months (Vd; hazard ratio, 0.76; 95% confidence interval, 0.633-0.915). OS was longer with Kd56 versus Vd within age and cytogenetic subgroups, and according to number of previous lines of therapy, previous bortezomib exposure, previous lenalidomide exposure, and lenalidomide-refractory status. Exposure-adjusted incidences per 100 patient-years of adverse events (AEs) were 1352.07 for Kd56 and 1754.86 for Vd; for Grade ≥3 AEs, these values were 162.31 and 175.90., Conclusion: With median follow-up of approximately 44 months, clinically meaningful improvements in OS were observed with Kd56 versus Vd, including in all subgroups examined. The Kd56 safety profile was consistent with previous analyses., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
20. Postoperative Radiotherapy for Multiple Myeloma of Long Bones: Should the Entire Rod Be Treated?
- Author
-
Elhammali A, Milgrom SA, Amini B, Gunther JR, Yoder A, Ludmir EB, Moon B, Weber DM, Thomas SK, Garg N, Manasanch EE, Patel KK, Orlowski RZ, Lee HC, Bird JE, Satcher R, Lin P, Pinnix CC, and Dabaja BS
- Subjects
- Aged, Aged, 80 and over, Female, Follow-Up Studies, Humans, Male, Middle Aged, Multiple Myeloma pathology, Multiple Myeloma surgery, Plasmacytoma pathology, Plasmacytoma surgery, Prognosis, Radiotherapy Dosage, Retrospective Studies, Survival Rate, Multiple Myeloma radiotherapy, Neoplasm Recurrence, Local prevention & control, Plasmacytoma radiotherapy, Postoperative Care
- Abstract
Purpose: To characterize local relapse after surgical fixation and postoperative radiotherapy (RT) for multiple myeloma (MM) with cortical involvement of long bones., Patients and Methods: We retrospectively identified patients with MM involving cortical long bones treated with surgical fixation followed by postoperative RT at our institution. Local failures, defined as radiographic recurrence along the surgical hardware, were documented, and potential associations of independent variables (RT dose, fractionation, and extent of hardware coverage) with local failure were assessed by univariate Cox regression., Results: We identified 33 patients with 40 treated sites with a median follow-up of 25.7 months; 68% of treatments were for pathologic fracture, and 32% were for impending fracture. The most common dose and fractionation were 20 to 25 Gy in 8 to 12 fractions. On average, 76% of the surgical hardware was covered by the postoperative RT field (median, 80%; range, 28%-100%). Local failure was observed in 5 cases (12.5%), 2 within the RT field and 3 out of field. None of the relapses resulted in hardware failure, and 2 were retreated with RT. The extent of hardware coverage predicted disease relapse along the hardware (hazard ratio = 6.44; 95% confidence interval, 1.09-37.97; P = .04); however, total RT dose, biologically effective dose, and number of fractions did not., Conclusion: After internal fixation of long bones with MM, full hardware coverage with the RT field could reduce the risk, though small, of disease developing in the future in the proximate hardware. Postoperative RT doses of 20 to 25 Gy in 8 to 10 fractions can achieve excellent local control., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
21. PD-L1 gene alterations identify a subset of diffuse large B-cell lymphoma harboring a T-cell-inflamed phenotype.
- Author
-
Godfrey J, Tumuluru S, Bao R, Leukam M, Venkataraman G, Phillip J, Fitzpatrick C, McElherne J, MacNabb BW, Orlowski R, Smith SM, and Kline J
- Subjects
- Adult, Disease-Free Survival, Female, Gene Expression Regulation, Neoplastic immunology, Humans, In Situ Hybridization, Fluorescence, Middle Aged, NF-kappa B genetics, NF-kappa B immunology, Signal Transduction genetics, Signal Transduction immunology, Survival Rate, B7-H1 Antigen genetics, B7-H1 Antigen immunology, Lymphoma, Large B-Cell, Diffuse genetics, Lymphoma, Large B-Cell, Diffuse immunology, Lymphoma, Large B-Cell, Diffuse mortality, Lymphoma, Large B-Cell, Diffuse therapy, Neoplasm Proteins genetics, Neoplasm Proteins immunology, T-Lymphocytes immunology, T-Lymphocytes pathology
- Abstract
Programmed death-ligand 1 (PD-L1) expression on malignant cells is a dominant immune escape mechanism across a variety of human cancers. A unique genetic mechanism underlying PD-L1 upregulation has been uncovered in classical Hodgkin lymphoma (cHL), in which copy gains of the chromosomal region (9p24.1) containing the programmed death-1 (PD-1) ligands PD-L1 and PD-L2 are recurrently observed. While chromosome 9p24.1 copy-number alterations are ubiquitous in cHL, they also occur in diffuse large B-cell lymphoma (DLBCL), albeit with a lower incidence. Here, fluorescence in situ hybridization was used to identify DLBCLs harboring PD-L1 gene alterations, thereby enabling a characterization of the immunogenomic landscape of these lymphomas. Among 105 DLBCL cases analyzed, PD-L1 alterations were identified in 27%. PD-L1 alterations were highly enriched among non-germinal center DLBCLs and exhibited robust PD-L1 protein expression. These lymphomas were heavily infiltrated by clonally restricted T cells and frequently downregulated human leukocyte antigen expression. RNA sequencing of PD-L1 -altered DLBCLs revealed upregulation of genes involved in negative T-cell regulation and NF-κB pathway activation, while whole-exome sequencing identified frequent mutations in genes involved in antigen presentation and T-cell costimulation. Many of these findings were validated in a large external data set. Interestingly, DLBCL patients with PD-L1 alterations had inferior progression-free survival following front-line chemoimmunotherapy; however, in the relapsed/refractory setting, PD-L1 alterations were associated with response to anti-PD-1 therapy. Collectively, our results indicate that PD-L1 alterations identify a unique biological subset of DLBCL in which an endogenous antilymphoma immune response has been activated, and that is associated with responsiveness to PD-1 blockade therapy., (© 2019 by The American Society of Hematology.)
- Published
- 2019
- Full Text
- View/download PDF
22. Ubiquitin-activating enzyme inhibition induces an unfolded protein response and overcomes drug resistance in myeloma.
- Author
-
Zhuang J, Shirazi F, Singh RK, Kuiatse I, Wang H, Lee HC, Berkova Z, Berger A, Hyer M, Chattopadhyay N, Syed S, Shi JQ, Yu J, Shinde V, Tirrell S, Jones RJ, Wang Z, Davis RE, and Orlowski RZ
- Subjects
- Antineoplastic Agents therapeutic use, Cell Line, Tumor, Drug Synergism, Endoplasmic Reticulum Stress drug effects, Humans, Oxidative Stress drug effects, Salvage Therapy methods, Tumor Cells, Cultured, Tumor Suppressor Protein p53 drug effects, Tumor Suppressor Protein p53 metabolism, Drug Resistance, Neoplasm drug effects, Multiple Myeloma drug therapy, Proteasome Inhibitors pharmacology, Ubiquitin-Activating Enzymes antagonists & inhibitors, Unfolded Protein Response drug effects
- Abstract
Three proteasome inhibitors have garnered regulatory approvals in various multiple myeloma settings; but drug resistance is an emerging challenge, prompting interest in blocking upstream components of the ubiquitin-proteasome pathway. One such attractive target is the E1 ubiquitin-activating enzyme (UAE); we therefore evaluated the activity of TAK-243, a novel and specific UAE inhibitor. TAK-243 potently suppressed myeloma cell line growth, induced apoptosis, and activated caspases while decreasing the abundance of ubiquitin-protein conjugates. This was accompanied by stabilization of many short-lived proteins, including p53, myeloid cell leukemia 1 (MCL-1), and c-MYC, and activation of the activating transcription factor 6 (ATF-6), inositol-requiring enzyme 1 (IRE-1), and protein kinase RNA-like endoplasmic reticulum (ER) kinase (PERK) arms of the ER stress response pathway, as well as oxidative stress. UAE inhibition showed comparable activity against otherwise isogenic cell lines with wild-type (WT) or deleted p53 despite induction of TP53 signaling in WT cells. Notably, TAK-243 overcame resistance to conventional drugs and novel agents in cell-line models, including bortezomib and carfilzomib resistance, and showed activity against primary cells from relapsed/refractory myeloma patients. In addition, TAK-243 showed strong synergy with a number of antimyeloma agents, including doxorubicin, melphalan, and panobinostat as measured by low combination indices. Finally, TAK-243 was active against a number of in vivo myeloma models in association with activation of ER stress. Taken together, the data support the conclusion that UAE inhibition could be an attractive strategy to move forward to the clinic for patients with relapsed and/or refractory multiple myeloma., (© 2019 by The American Society of Hematology.)
- Published
- 2019
- Full Text
- View/download PDF
23. The Imipridone ONC201 Induces Apoptosis and Overcomes Chemotherapy Resistance by Up-Regulation of Bim in Multiple Myeloma.
- Author
-
Tu YS, He J, Liu H, Lee HC, Wang H, Ishizawa J, Allen JE, Andreeff M, Orlowski RZ, Davis RE, and Yang J
- Subjects
- Bcl-2-Like Protein 11 metabolism, Cell Line, Tumor, Cell Survival drug effects, Dose-Response Relationship, Drug, Drug Synergism, Gene Expression Regulation, Neoplastic drug effects, Humans, Imidazoles, Models, Biological, Multiple Myeloma drug therapy, Pyridines, Pyrimidines, RNA Interference, RNA, Small Interfering genetics, Signal Transduction drug effects, Antineoplastic Agents pharmacology, Apoptosis drug effects, Bcl-2-Like Protein 11 genetics, Drug Resistance, Neoplasm drug effects, Heterocyclic Compounds, 4 or More Rings pharmacology, Multiple Myeloma genetics, Multiple Myeloma metabolism
- Abstract
In multiple myeloma, despite recent improvements offered by new therapies, disease relapse and drug resistance still occur in the majority of patients. Therefore, there is an urgent need for new drugs that can overcome drug resistance and prolong patient survival after failure of standard therapies. The imipridone ONC201 causes downstream inactivation of ERK1/2 signaling and has tumoricidal activity against a variety of tumor types, while its efficacy in preclinical models of myeloma remains unclear. In this study, we treated human myeloma cell lines and patient-derived tumor cells with ONC201. Treatment decreased cellular viability and induced apoptosis in myeloma cell lines, with IC50 values of 1 to 1.5 μM, even in those with high risk features or TP53 loss. ONC201 increased levels of the pro-apoptotic protein Bim in myeloma cells, resulting from decreased phosphorylation of degradation-promoting Bim Ser69 by ERK1/2. In addition, myeloma cell lines made resistant to several standard-of-care agents (by chronic exposure) were equally sensitive to ONC201 as their drug-naïve counterparts, and combinations of ONC201 with proteasome inhibitors had synergistic anti-myeloma activity. Overall, these findings demonstrate that ONC201 kills myeloma cells regardless of resistance to standard-of-care therapies, making it promising for clinical testing in relapsed/refractory myeloma., (Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
24. Final Results of a Phase 1 Study of Vorinostat, Pegylated Liposomal Doxorubicin, and Bortezomib in Relapsed or Refractory Multiple Myeloma.
- Author
-
Voorhees PM, Gasparetto C, Moore DT, Winans D, Orlowski RZ, and Hurd DD
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols pharmacology, Bortezomib administration & dosage, Bortezomib pharmacology, Doxorubicin administration & dosage, Doxorubicin pharmacology, Doxorubicin therapeutic use, Drug Resistance, Neoplasm, Female, Humans, Hydroxamic Acids administration & dosage, Hydroxamic Acids pharmacology, Male, Middle Aged, Multiple Myeloma drug therapy, Multiple Myeloma pathology, Neoplasm Recurrence, Local, Polyethylene Glycols administration & dosage, Polyethylene Glycols pharmacology, Polyethylene Glycols therapeutic use, Proteasome Inhibitors administration & dosage, Proteasome Inhibitors pharmacology, Vorinostat, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bortezomib therapeutic use, Doxorubicin analogs & derivatives, Hydroxamic Acids therapeutic use, Proteasome Inhibitors therapeutic use
- Abstract
Introduction/background: Deacetylase inhibitors have synergistic activity in combination with proteasome inhibitors and anthracyclines in preclinical models of multiple myeloma (MM). We therefore evaluated the safety and efficacy of the deacetylase inhibitor vorinostat in combination with pegylated liposomal doxorubicin (PLD) and bortezomib in relapsed/refractory MM., Patients and Methods: Thirty-two patients were treated with PLD and bortezomib in combination with escalating doses of vorinostat on days 4 to 11 or 1 to 14., Results: The maximum tolerated dose of vorinostat was 400 mg on days 4 to 11. Neutropenia and thrombocytopenia attributable to protocol therapy were seen in 59% and 94% of patients, of which 37% and 47% were of grade 3 or higher severity, respectively. Constitutional and gastrointestinal adverse events of all grades were common, the majority of which were less than grade 3 in severity. The overall response rate (partial response rate or better) was 65% and the clinical benefit rate (minimal response rate or better) 74%. The overall response rate was 83%, 71%, and 45% for patients with bortezomib-naive, -sensitive, and -refractory MM, respectively. The median progression-free survival was 13.9 months and the 3-year overall survival 77%. Whole blood proteasome activity assays demonstrated a potential impact of vorinostat on the chymotryptic-like activity of the proteasome., Conclusion: Further evaluation of PLD, bortezomib, and deacetylase inhibitor combinations is warranted, with special attention directed toward strategies to improve tolerability., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
25. Clinical and pathological characteristics of HIV- and HHV-8-negative Castleman disease.
- Author
-
Yu L, Tu M, Cortes J, Xu-Monette ZY, Miranda RN, Zhang J, Orlowski RZ, Neelapu S, Boddu PC, Akosile MA, Uldrick TS, Yarchoan R, Medeiros LJ, Li Y, Fajgenbaum DC, and Young KH
- Subjects
- Adolescent, Adult, Aged, Antibodies, Monoclonal therapeutic use, Castleman Disease pathology, Castleman Disease virology, Disease-Free Survival, Female, HIV-1, Herpesvirus 8, Human, Humans, Immunophenotyping, Inflammation, Male, Middle Aged, Young Adult, Castleman Disease therapy
- Abstract
Castleman disease (CD) comprises 3 poorly understood lymphoproliferative variants sharing several common histopathological features. Unicentric CD (UCD) is localized to a single region of lymph nodes. Multicentric CD (MCD) manifests with systemic inflammatory symptoms and organ dysfunction due to cytokine dysregulation and involves multiple lymph node regions. Human herpesvirus 8 (HHV-8) causes MCD (HHV-8-associated MCD) in immunocompromised individuals, such as HIV-infected patients. However, >50% of MCD cases are HIV and HHV-8 negative (defined as idiopathic [iMCD]). The clinical and biological behavior of CD remains poorly elucidated. Here, we analyzed the clinicopathologic features of 74 patients (43 with UCD and 31 with iMCD) and therapeutic response of 96 patients (43 with UCD and 53 with iMCD) with HIV-/HHV-8-negative CD compared with 51 HIV-/HHV-8-positive patients. Systemic inflammatory symptoms and elevated inflammatory factors were more common in iMCD patients than UCD patients. Abnormal bone marrow features were more frequent in iMCD (77.0%) than UCD (45%); the most frequent was plasmacytosis, which was seen in 3% to 30.4% of marrow cells. In the lymph nodes, higher numbers of CD3
+ lymphocytes (median, 58.88 ± 20.57) and lower frequency of CD19+ /CD5+ (median, 5.88 ± 6.52) were observed in iMCD patients compared with UCD patients (median CD3+ cells, 43.19 ± 17.37; median CD19+ /CD5+ cells, 17.37 ± 15.80). Complete surgical resection is a better option for patients with UCD. Siltuximab had a greater proportion of complete responses and longer progression-free survival (PFS) for iMCD than rituximab. Centricity, histopathological type, and anemia significantly impacted PFS. This study reveals that CD represents a heterogeneous group of diseases with differential immunophenotypic profiling and treatment response.- Published
- 2017
- Full Text
- View/download PDF
26. Bortezomib with lenalidomide and dexamethasone versus lenalidomide and dexamethasone alone in patients with newly diagnosed myeloma without intent for immediate autologous stem-cell transplant (SWOG S0777): a randomised, open-label, phase 3 trial.
- Author
-
Durie BGM, Hoering A, Abidi MH, Rajkumar SV, Epstein J, Kahanic SP, Thakuri M, Reu F, Reynolds CM, Sexton R, Orlowski RZ, Barlogie B, and Dispenzieri A
- Subjects
- Adult, Aged, Disease-Free Survival, Drug Therapy, Combination, Female, Hematopoietic Stem Cell Transplantation, Humans, Lenalidomide, Male, Middle Aged, Multiple Myeloma mortality, Survival Rate, Thalidomide therapeutic use, Treatment Outcome, Angiogenesis Inhibitors therapeutic use, Antineoplastic Agents therapeutic use, Bortezomib therapeutic use, Dexamethasone therapeutic use, Multiple Myeloma drug therapy, Thalidomide analogs & derivatives
- Abstract
Background: Lenalidomide plus dexamethasone is a reference treatment for patients with newly diagnosed myeloma. The combination of the proteasome inhibitor bortezomib with lenalidomide and dexamethasone has shown significant efficacy in the setting of newly diagnosed myeloma. We aimed to study whether the addition of bortezomib to lenalidomide and dexamethasone would improve progression-free survival and provide better response rates in patients with previously untreated multiple myeloma who were not planned for immediate autologous stem-cell transplant., Methods: In this randomised, open-label, phase 3 trial, we recruited patients with newly diagnosed multiple myeloma aged 18 years and older from participating Southwest Oncology Group (SWOG) and National Clinical Trial Network (NCTN) institutions (both inpatient and outpatient settings). Key inclusion criteria were presence of CRAB (C=calcium elevation; R=renal impairment; A=anaemia; B=bone involvement) criteria with measurable disease (measured by assessment of free light chains), Eastern Cooperative Oncology Group (ECOG) performance status of 0-3, haemoglobin concentration 9 g/dL or higher, absolute neutrophil count 1 × 10
3 cells per mm3 or higher, and a platelet count of 80 000/mm3 or higher. We randomly assigned (1:1) patients to receive either an initial treatment of bortezomib with lenalidomide and dexamethasone (VRd group) or lenalidomide and dexamethasone alone (Rd group). Randomisation was stratified based on International Staging System stage (I, II, or III) and intent to transplant (yes vs no). The VRd regimen was given as eight 21-day cycles. Bortezomib was given at 1·3 mg/m2 intravenously on days 1, 4, 8, and 11, combined with oral lenalidomide 25 mg daily on days 1-14 plus oral dexamethasone 20 mg daily on days 1, 2, 4, 5, 8, 9, 11, and 12. The Rd regimen was given as six 28-day cycles. The standard Rd regimen consisted of 25 mg oral lenalidomide once a day for days 1-21 plus 40 mg oral dexamethasone once a day on days 1, 8, 15, and 22. The primary endpoint was progression-free survival using a prespecified one-sided stratified log rank test at a significance level of 0·02. Analyses were intention to treat. This trial is registered with ClinicalTrials.gov, number NCT00644228., Findings: Between April, 2008, and February, 2012, we randomly assigned 525 patients at 139 participating institutions (264 to VRd and 261 to Rd). In the randomly assigned patients, 21 patients in the VRd group and 31 in the Rd group were deemed ineligible based mainly on missing, insufficient, or early or late baseline laboratory data. Median progression-free survival was significantly improved in the VRd group (43 months vs 30 months in the Rd group; stratified hazard ratio [HR] 0·712, 96% CI 0·56-0·906; one-sided p value 0·0018). The median overall survival was also significantly improved in the VRd group (75 months vs 64 months in the Rd group, HR 0·709, 95% CI 0·524-0·959; two-sided p value 0·025). The rates of overall response (partial response or better) were 82% (176/216) in the VRd group and 72% (153/214) in the Rd group, and 16% (34/216) and 8% (18/214) of patients who were assessable for response in these respective groups had a complete response or better. Adverse events of grade 3 or higher were reported in 198 (82%) of 241 patients in the VRd group and 169 (75%) of 226 patients in the Rd group; 55 (23%) and 22 (10%) patients discontinued induction treatment because of adverse events, respectively. There were no treatment-related deaths in the Rd group, and two in the VRd group., Interpretation: In patients with newly diagnosed myeloma, the addition of bortezomib to lenalidomide and dexamethasone resulted in significantly improved progression-free and overall survival and had an acceptable risk-benefit profile., Funding: NIH, NCI, NCTN, Millennium Pharmaceuticals, Takeda Oncology Company, and Celgene Corporation., (Copyright © 2017 Elsevier Ltd. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
27. Treatment of multiple myeloma with high-risk cytogenetics: a consensus of the International Myeloma Working Group.
- Author
-
Sonneveld P, Avet-Loiseau H, Lonial S, Usmani S, Siegel D, Anderson KC, Chng WJ, Moreau P, Attal M, Kyle RA, Caers J, Hillengass J, San Miguel J, van de Donk NW, Einsele H, Bladé J, Durie BG, Goldschmidt H, Mateos MV, Palumbo A, and Orlowski R
- Subjects
- Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bortezomib, Chromosome Aberrations classification, Combined Modality Therapy, Consensus, Cytogenetics, Hematopoietic Stem Cell Transplantation, Humans, Lenalidomide, Multiple Myeloma classification, Multiple Myeloma genetics, Multiple Myeloma mortality, Prognosis, Risk Factors, Thalidomide analogs & derivatives, Transplantation, Autologous, Multiple Myeloma therapy
- Abstract
The International Myeloma Working Group consensus updates the definition for high-risk (HR) multiple myeloma based on cytogenetics Several cytogenetic abnormalities such as t(4;14), del(17/17p), t(14;16), t(14;20), nonhyperdiploidy, and gain(1q) were identified that confer poor prognosis. The prognosis of patients showing these abnormalities may vary with the choice of therapy. Treatment strategies have shown promise for HR cytogenetic diseases, such as proteasome inhibition in combination with lenalidomide/pomalidomide, double autologous stem cell transplant plus bortezomib, or combination of immunotherapy with lenalidomide or pomalidomide. Careful analysis of cytogenetic subgroups in trials comparing different treatments remains an important goal. Cross-trial comparisons may provide insight into the effect of new drugs in patients with cytogenetic abnormalities. However, to achieve this, consensus on definitions of analytical techniques, proportion of abnormal cells, and treatment regimens is needed. Based on data available today, bortezomib and carfilzomib treatment appear to improve complete response, progression-free survival, and overall survival in t(4;14) and del(17/17p), whereas lenalidomide may be associated with improved progression-free survival in t(4;14) and del(17/17p). Patients with multiple adverse cytogenetic abnormalities do not benefit from these agents. FISH data are implemented in the revised International Staging System for risk stratification., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
28. Daratumumab monotherapy in patients with treatment-refractory multiple myeloma (SIRIUS): an open-label, randomised, phase 2 trial.
- Author
-
Lonial S, Weiss BM, Usmani SZ, Singhal S, Chari A, Bahlis NJ, Belch A, Krishnan A, Vescio RA, Mateos MV, Mazumder A, Orlowski RZ, Sutherland HJ, Bladé J, Scott EC, Oriol A, Berdeja J, Gharibo M, Stevens DA, LeBlanc R, Sebag M, Callander N, Jakubowiak A, White D, de la Rubia J, Richardson PG, Lisby S, Feng H, Uhlar CM, Khan I, Ahmadi T, and Voorhees PM
- Subjects
- Adult, Aged, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal adverse effects, Antineoplastic Agents administration & dosage, Antineoplastic Agents adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Canada, Disease-Free Survival, Drug Administration Schedule, Drug Resistance, Neoplasm, Female, Humans, Kaplan-Meier Estimate, Male, Middle Aged, Spain, Treatment Outcome, United States, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents therapeutic use, Multiple Myeloma drug therapy, Neoplasm Recurrence, Local drug therapy
- Abstract
Background: New treatment options are needed for patients with multiple myeloma that is refractory to proteasome inhibitors and immunomodulatory drugs. We assessed daratumumab, a novel CD38-targeted monoclonal antibody, in patients with refractory multiple myeloma., Methods: In this open-label, multicentre, phase 2 trial done in Canada, Spain, and the USA, patients (age ≥18 years) with multiple myeloma who were previously treated with at least three lines of therapy (including proteasome inhibitors and immunomodulatory drugs), or were refractory to both proteasome inhibitors and immunomodulatory drugs, were randomly allocated in a 1:1 ratio to receive intravenous daratumumab 8 mg/kg or 16 mg/kg in part 1 stage 1 of the study, to decide the dose for further assessment in part 2. Patients received 8 mg/kg every 4 weeks, or 16 mg/kg per week for 8 weeks (cycles 1 and 2), then every 2 weeks for 16 weeks (cycles 3-6), and then every 4 weeks thereafter (cycle 7 and higher). The allocation schedule was computer-generated and randomisation, with permuted blocks, was done centrally with an interactive web response system. In part 1 stage 2 and part 2, patients received 16 mg/kg dosed as in part 1 stage 1. The primary endpoint was overall response rate (partial response [PR] + very good PR + complete response [CR] + stringent CR). All patients who received at least one dose of daratumumab were included in the analysis. The trial is registered with ClinicalTrials.gov, number NCT01985126., Findings: The study is ongoing. In part 1 stage 1 of the study, 18 patients were randomly allocated to the 8 mg/kg group and 16 to the 16 mg/kg group. Findings are reported for the 106 patients who received daratumumab 16 mg/kg in parts 1 and 2. Patients received a median of five previous lines of therapy (range 2-14). 85 (80%) patients had previously received autologous stem cell transplantation, 101 (95%) were refractory to the most recent proteasome inhibitors and immunomodulatory drugs used, and 103 (97%) were refractory to the last line of therapy. Overall responses were noted in 31 patients (29.2%, 95% CI 20.8-38.9)-three (2.8%, 0.6-8.0) had a stringent CR, ten (9.4%, 4.6-16.7) had a very good PR, and 18 (17.0%, 10.4-25.5) had a PR. The median time to first response was 1.0 month (range 0.9-5.6). Median duration of response was 7.4 months (95% CI 5.5-not estimable) and progression-free survival was 3.7 months (95% CI 2.8-4.6). The 12-month overall survival was 64.8% (95% CI 51.2-75.5) and, at a subsequent cutoff, median overall survival was 17.5 months (95% CI 13.7-not estimable). Daratumumab was well tolerated; fatigue (42 [40%] patients) and anaemia (35 [33%]) of any grade were the most common adverse events. No drug-related adverse events led to treatment discontinuation., Interpretation: Daratumumab monotherapy showed encouraging efficacy in heavily pretreated and refractory patients with multiple myeloma, with a favourable safety profile in this population of patients., Funding: Janssen Research & Development., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
29. Outcome of Patients With Nonsecretory Multiple Myeloma After Autologous Hematopoietic Stem Cell Transplantation.
- Author
-
Jacobs RW, Saliba RM, Sasaki K, Farhan S, Armas A, Shah ND, Bashir Q, Qureshi S, Rondon G, Hosing C, Popat U, Parmar S, Shah JJ, Wang M, Weber DM, Thomas SK, Orlowski RZ, Champlin RE, and Qazilbash MH
- Subjects
- Adult, Aged, Disease-Free Survival, Female, Humans, Induction Chemotherapy, Male, Middle Aged, Multiple Myeloma mortality, Multiple Myeloma pathology, Myeloma Proteins analysis, Neoplasm Staging, Recurrence, Transplantation Conditioning, Treatment Outcome, Hematopoietic Stem Cell Transplantation, Multiple Myeloma surgery, Transplantation, Autologous
- Abstract
Introduction: Fewer than 5% of patients have nonsecretory multiple myeloma (NSM), which is characterized by the absence of monoclonal protein in immunofixation in serum and urine. There are limited data on the outcome of NSM after autologous hematopoietic stem cell transplantation (auto-HCT)., Patients and Methods: Between 1988 and December 2010, we identified 31 patients with NSM, and compared their outcome with 124 patients with secretory myeloma (SM) who were matched for age, disease stage, year of auto-HCT, and disease status and received auto-HCT at our institution. The primary end points were time to progression (TTP), progression-free survival (PFS), and overall survival (OS)., Results: Nonrelapse mortality at 4 years was 4% and 4% in the NSM and SM patients, respectively (P = .612). Median follow-up was 102 and 74 months for NSM and SM patients, respectively. Median PFS was 37 (95% confidence interval [CI], 12-62) and 22 (95% CI, 18-26) months for NSM and SM patients, respectively (P = .527). Median OS was 51 (95% CI, 7-95) and 73 (95% CI, 59-86) months for NSM and SM patients, respectively (P = .740). In multivariate analyses, age >55 years, and relapsed disease were associated with a shorter TTP. Similarly, age >55 years, and relapsed or refractory disease at the time of auto-HCT were associated with a shorter OS., Conclusion: Auto-HCT is associated with durable remission in NSM. There was no significant difference in transplant-related mortality, TTP, and PFS in patients with NSM compared with patients with SM after high-dose therapy and auto-HCT., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
30. Prospective analysis of antigen-specific immunity, stem-cell antigens, and immune checkpoints in monoclonal gammopathy.
- Author
-
Dhodapkar MV, Sexton R, Das R, Dhodapkar KM, Zhang L, Sundaram R, Soni S, Crowley JJ, Orlowski RZ, and Barlogie B
- Subjects
- B-Lymphocytes pathology, B7-H1 Antigen immunology, Female, Humans, Male, Multiple Myeloma pathology, Multiple Myeloma prevention & control, Prospective Studies, Stem Cells pathology, T-Lymphocytes pathology, Antigens, Neoplasm immunology, B-Lymphocytes immunology, Immunity, Cellular, Multiple Myeloma immunology, SOXB1 Transcription Factors immunology, Stem Cells immunology, T-Lymphocytes immunology
- Abstract
Blockade of immune checkpoints (ICPs) has led to impressive responses in cancer patients. However, the impact of preexisting immunity and ICPs on the risk of malignant transformation in human preneoplasia has not been prospectively studied. We prospectively analyzed antigen-specific B/T-cell immunity, immune composition of the tumor microenvironment, and the expression of a panel of ICPs on tumor and tumor-infiltrating immune cells in 305 patients with asymptomatic monoclonal gammopathy enrolled in S0120 under the auspices of SWOG. T-cell immunity against stem-cell antigen SOX2 and preserved humoral responses at study entry independently correlated with reduced risk of progression to clinical myeloma. Among the ICPs analyzed, expression of programmed death-ligand 1 (PD-L1) on tumor and infiltrating T cells correlated with increased risk of clinical malignancy, and blockade of this pathway boosted anti-SOX2 immunity in culture. These data suggest that stem-cell antigens and PD-L1 may be targeted for immunoprevention of myeloma. This trial was registered at www.clinicaltrials.gov as #NCT00900263., (© 2015 by The American Society of Hematology.)
- Published
- 2015
- Full Text
- View/download PDF
31. Carfilzomib, pomalidomide, and dexamethasone for relapsed or refractory myeloma.
- Author
-
Shah JJ, Stadtmauer EA, Abonour R, Cohen AD, Bensinger WI, Gasparetto C, Kaufman JL, Lentzsch S, Vogl DT, Gomes CL, Pascucci N, Smith DD, Orlowski RZ, and Durie BG
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Dexamethasone administration & dosage, Dexamethasone adverse effects, Female, Humans, Male, Maximum Tolerated Dose, Middle Aged, Multiple Myeloma mortality, Multiple Myeloma pathology, Oligopeptides administration & dosage, Oligopeptides adverse effects, Pneumonia chemically induced, Pneumonia mortality, Pulmonary Embolism chemically induced, Pulmonary Embolism mortality, Recurrence, Thalidomide administration & dosage, Thalidomide adverse effects, Thalidomide analogs & derivatives, Time Factors, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Multiple Myeloma drug therapy
- Abstract
Treatment options for patients with heavily pretreated relapsed and/or refractory multiple myeloma remain limited. We evaluated a novel therapeutic regimen consisting of carfilzomib, pomalidomide, and dexamethasone (CPD) in an open-label, multicenter, phase 1, dose-escalation study. Patients who relapsed after prior therapy or were refractory to the most recently received therapy were eligible. All patients were refractory to prior lenalidomide. Patients received carfilzomib IV on days 1, 2, 8, 9, 15, and 16 (starting dose of 20/27 mg/m(2)), pomalidomide once daily on days 1 to 21 (4 mg as the initial dose level), and dexamethasone (40 mg oral or IV) on days 1, 8, 15, and 22 of 28-day cycles. The primary objective was to evaluate the safety and determine the maximum tolerated dose (MTD) of the regimen. A total of 32 patients were enrolled. The MTD of the regimen was dose level 1 (carfilzomib 20/27 mg/m(2), pomalidomide 4 mg, dexamethasone 40 mg). Hematologic adverse events (AEs) occurred in ≥60% of all patients, including 11 patients with grade ≥3 anemia. Dyspnea was limited to grade 1/2 in 10 patients. Peripheral neuropathy was uncommon and limited to grade 1/2. Eight patients had dose reductions during therapy, and 7 patients discontinued treatment due to AEs. Two deaths were noted on study due to pneumonia and pulmonary embolism (n = 1 each). The combination of CPD is well-tolerated and highly active in patients with relapsed/refractory multiple myeloma. This trial was registered at www.clinicaltrials.gov as #NCT01464034., (© 2015 by The American Society of Hematology.)
- Published
- 2015
- Full Text
- View/download PDF
32. Outcomes Among High-Risk and Standard-Risk Multiple Myeloma Patients Treated With High-Dose Chemotherapy and Autologous Hematopoietic Stem-Cell Transplantation.
- Author
-
Kazmi SM, Nusrat M, Gunaydin H, Cornelison AM, Shah N, Kebriaei P, Nieto Y, Parmar S, Popat UR, Oran B, Shah JJ, Orlowski RZ, Champlin RE, Qazilbash MH, and Bashir Q
- Subjects
- Adult, Aged, Aged, 80 and over, Biomarkers, Female, Graft Survival, Humans, Male, Middle Aged, Multiple Myeloma diagnosis, Multiple Myeloma mortality, Neoplasm Staging, Survival Analysis, Transplantation Conditioning, Transplantation, Autologous, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Chromosome Aberrations, Hematopoietic Stem Cell Transplantation adverse effects, Multiple Myeloma genetics, Multiple Myeloma therapy
- Abstract
Background: Conventional cytogenetics and interphase fluorescence in-situ hybridization (FISH) identify a high-risk multiple myeloma population characterized by poor response and short survival., Patients and Methods: We compared outcomes between high-risk and standard-risk myeloma patients who underwent autologous hematopoietic stem-cell transplantation (auto-HCT) at our institution between January 2005 and December 2009. High-risk myeloma was defined as -13/del(13q) or hypodiploidy in at least 2 metaphases of conventional cytogenetics, or -17/del(17p), t(4;14), t(14;16), t(14;20), hypodiploidy (< 45 chromosomes excluding -Y), or chromosome 1 abnormalities (+1q, -1p, t(1;x)) on FISH or conventional cytogenetics., Results: Of 670 myeloma patients, 74 (11%) had high-risk myeloma. These high-risk patients had significantly lower overall response rates (74% vs. 85%; P < .01), shorter median progression-free survival (10.3 vs. 32.4 months; P < .001), and shorter overall survival (28 months vs. not reached; P < .001) than the standard-risk patients. Having only 1 high-risk cytogenetic abnormality or experiencing at least very good partial remission after auto-HCT independently predicted improved progression-free survival and overall survival (P < .05) in high-risk patients., Conclusion: Even in an era of novel therapies, cytogenetically identified high-risk myeloma patients have worse prognoses than standard-risk myeloma patients after auto-HCT, and having more than 1 high-risk cytogenetic abnormality further reduces survival., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
33. Autologous Hematopoietic Stem Cell Transplantation in Dialysis-Dependent Myeloma Patients.
- Author
-
El Fakih R, Fox P, Popat U, Nieto Y, Shah N, Parmar S, Oran B, Ciurea S, Kebriaei P, Hosing C, Ahmed S, Shah J, Orlowski R, Champlin R, Qazilbash M, and Bashir Q
- Subjects
- Adult, Aged, Disease-Free Survival, Female, Humans, Incidence, Male, Middle Aged, Retrospective Studies, Hematopoietic Stem Cell Transplantation methods, Multiple Myeloma surgery, Multiple Myeloma therapy, Transplantation Conditioning methods, Transplantation, Autologous methods
- Abstract
Background: We retrospectively analyzed our transplant database from July 2000 to June 2012 to identify myeloma patients who received autologous stem cell transplantation while dialysis-dependent., Patients: 2091 patients underwent autologous high-dose therapy during this period. Twenty-four patients were dialysis-dependent., Results: The 100-day and the 6, and 12-month treatment-related mortality was 0%. Overall response rate was 92%. The median progression-free survival and overall survival were 1.9 years and 3.8 years, respectively. A multivariate analysis was not performed because of the small sample size. Only 3 patients became dialysis-independent after transplantation. Cardiac, gastrointestinal, genitourinary, infectious, neurologic, and pulmonary "all grade" toxicities were all higher in the melphalan 200 group versus < 200 group, however, none of them were statistically significant., Conclusion: Because of a lack of clear survival benefit with higher-dose melphalan and potential higher toxicity in this group, it is reasonable to use lower-dose melphalan in dialysis-dependent myeloma patients., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
34. Targeting the pro-survival protein MET with tivantinib (ARQ 197) inhibits growth of multiple myeloma cells.
- Author
-
Zaman S, Shentu S, Yang J, He J, Orlowski RZ, Stellrecht CM, and Gandhi V
- Subjects
- Animals, Bortezomib pharmacology, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Dexamethasone pharmacology, Disease Models, Animal, Drug Resistance, Neoplasm, Humans, Lenalidomide, Melphalan pharmacology, Mice, Multiple Myeloma pathology, Signal Transduction drug effects, Thalidomide analogs & derivatives, Thalidomide pharmacology, Tumor Microenvironment drug effects, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Multiple Myeloma metabolism, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-met antagonists & inhibitors, Proto-Oncogene Proteins c-met metabolism, Pyrrolidinones pharmacology, Quinolines pharmacology
- Abstract
The hepatocyte growth factor (HGF)/MNNG HOS transforming gene (MET) pathway regulates cell growth, survival, and migration. MET is mutated or amplified in several malignancies. In myeloma, MET is not mutated, but patients have high plasma concentrations of HGF, high levels of MET expression, and gene copy number, which are associated with poor prognosis and advanced disease. Our previous studies demonstrated that MET is critical for myeloma cell survival and its knockdown induces apoptosis. In our current study, we tested tivantinib (ARQ 197), a small-molecule pharmacological MET inhibitor. At clinically achievable concentrations, tivantinib induced apoptosis by >50% in all 12 human myeloma cell lines tested. This biologic response was associated with down-regulation of MET signaling and inhibition of the mitogen-activated protein kinase and phosphoinositide 3-kinase pathways, which are downstream of the HGF/MET axis. Tivantinib was equally effective in inducing apoptosis in myeloma cell lines resistant to standard chemotherapy (melphalan, dexamethasone, bortezomib, and lenalidomide) as well as in cells that were co-cultured with a protective bone marrow microenvironment or with exogenous cytokines. Tivantinib induced apoptosis in CD138+ plasma cells from patients and demonstrated efficacy in a myeloma xenograft mouse model. On the basis of these data, we initiated a clinical trial for relapsed/refractory multiple myeloma (MM). In conclusion, MET inhibitors may be an attractive target-based strategy for the treatment of MM., (Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
35. Characteristics and outcomes of patients with multiple myeloma who develop therapy-related myelodysplastic syndrome, chronic myelomonocytic leukemia, or acute myeloid leukemia.
- Author
-
Pemmaraju N, Shah D, Kantarjian H, Orlowski RZ, Nogueras González GM, Baladandayuthapani V, Jain N, Wagner V, Garcia-Manero G, Shah J, Ravandi F, Pierce S, Takahashi K, Daver N, Nazha A, Verstovsek S, Jabbour E, De Lima M, Champlin R, Cortes J, and Qazilbash MH
- Subjects
- Aged, Aged, 80 and over, Female, Humans, Male, Middle Aged, Survival Rate, Treatment Outcome, Leukemia, Myeloid, Acute etiology, Leukemia, Myelomonocytic, Chronic etiology, Multiple Myeloma pathology, Multiple Myeloma therapy, Myelodysplastic Syndromes etiology, Neoplasms, Second Primary etiology
- Abstract
Background: Patients with multiple myeloma (MM) have had significant improvements in outcomes. An increased risk of therapy-related myeloid neoplasms (t-MNs) has also developed. Little is known about the characteristics and outcomes of these patients., Patients and Methods: Patients with MM treated at our institution from 1993 to 2011 were reviewed. Forty-seven patients were diagnosed with t-MN. Our primary objective was to evaluate the interval to t-MN, response to treatment, and overall survival (OS)., Results: The median patient age at the MM diagnosis was 65 years. Of the 47 patients, 32 (68.0%) initially received conventional chemotherapeutic agents, 7 (14.9%), novel agents (eg, lenalidomide, thalidomide, bortezomib), and 8 (17.0%), a combination. Twenty patients (42.6%) underwent high-dose chemotherapy and autologous hematopoietic stem cell transplantation. The median interval from the MM diagnosis to t-MN was 7 years (95% CI, 5.0-28.0). Of the 47 patients, 33 (70.2%) developed therapy-related myelodysplastic syndrome (t-MDS), 11 (23.4%) acute myeloid leukemia (t-AML), and 3 (6.4%) chronic myelomonocytic leukemia (t-CMML). The median age at the t-MN diagnosis was 65 years. Of the 47 patients, 26 (78.8%) with t-MDS, 9 (81.8%) with t-AML, and 1 (33.3%) with t-CMML had complex/high-risk cytogenetics. The median OS for all 47 patients after the t-MN diagnosis was 6.3 months (95% CI, 4.0-8.7)., Conclusion: The development of t-MN in patients with MM is associated with poor outcomes. These patients, in general, have complex cytogenetic abnormalities and short complete remission and OS times. A better understanding of the disease biology and novel therapeutic approaches are warranted., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
36. A Bim-targeting strategy overcomes adaptive bortezomib resistance in myeloma through a novel link between autophagy and apoptosis.
- Author
-
Chen S, Zhang Y, Zhou L, Leng Y, Lin H, Kmieciak M, Pei XY, Jones R, Orlowski RZ, Dai Y, and Grant S
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Apoptosis drug effects, Apoptosis genetics, Apoptosis Regulatory Proteins antagonists & inhibitors, Apoptosis Regulatory Proteins genetics, Autophagy drug effects, Autophagy genetics, Bcl-2-Like Protein 11, Boronic Acids pharmacology, Bortezomib, Cell Line, Tumor, Cells, Cultured, Drug Resistance, Neoplasm drug effects, Fluorescent Antibody Technique, Histone Deacetylase Inhibitors administration & dosage, Histone Deacetylase Inhibitors pharmacology, Humans, Hydroxamic Acids administration & dosage, Hydroxamic Acids pharmacology, Immunoblotting, Membrane Proteins antagonists & inhibitors, Membrane Proteins genetics, Mice, Inbred NOD, Mice, Knockout, Mice, SCID, Multiple Myeloma drug therapy, Multiple Myeloma genetics, Proto-Oncogene Proteins antagonists & inhibitors, Proto-Oncogene Proteins genetics, Pyrazines pharmacology, RNA Interference, Tumor Burden drug effects, Tumor Burden genetics, Xenograft Model Antitumor Assays, Apoptosis physiology, Apoptosis Regulatory Proteins metabolism, Autophagy physiology, Membrane Proteins metabolism, Multiple Myeloma metabolism, Proto-Oncogene Proteins metabolism
- Abstract
Bim contributes to resistance to various standard and novel agents. Here we demonstrate that Bim plays a functional role in bortezomib resistance in multiple myeloma (MM) cells and that targeting Bim by combining histone deacetylase inhibitors (HDACIs) with BH3 mimetics (eg, ABT-737) overcomes bortezomib resistance. BH3-only protein profiling revealed high Bim levels (Bim(hi)) in most MM cell lines and primary CD138(+) MM samples. Whereas short hairpin RNA Bim knockdown conferred bortezomib resistance in Bim(hi) cells, adaptive bortezomib-resistant cells displayed marked Bim downregulation. HDACI upregulated Bim and, when combined with ABT-737, which released Bim from Bcl-2/Bcl-xL, potently killed bortezomib-resistant cells. These events were correlated with Bim-associated autophagy attenuation, whereas Bim knockdown sharply increased autophagy in Bim(hi) cells. In Bim(low) cells, autophagy disruption by chloroquine (CQ) was required for HDACI/ABT-737 to induce Bim expression and lethality. CQ also further enhanced HDACI/ABT-737 lethality in bortezomib-resistant cells. Finally, HDACI failed to diminish autophagy or potentiate ABT-737-induced apoptosis in bim(-/-) mouse embryonic fibroblasts. Thus, Bim deficiency represents a novel mechanism of adaptive bortezomib resistance in MM cells, and Bim-targeting strategies combining HDACIs (which upregulate Bim) and BH3 mimetics (which unleash Bim from antiapoptotic proteins) overcomes such resistance, in part by disabling cytoprotective autophagy., (© 2014 by The American Society of Hematology.)
- Published
- 2014
- Full Text
- View/download PDF
37. Phase 2 randomized study of bortezomib-melphalan-prednisone with or without siltuximab (anti-IL-6) in multiple myeloma.
- Author
-
San-Miguel J, Bladé J, Shpilberg O, Grosicki S, Maloisel F, Min CK, Polo Zarzuela M, Robak T, Prasad SV, Tee Goh Y, Laubach J, Spencer A, Mateos MV, Palumbo A, Puchalski T, Reddy M, Uhlar C, Qin X, van de Velde H, Xie H, and Orlowski RZ
- Subjects
- Aged, Aged, 80 and over, Antibodies, Monoclonal administration & dosage, Boronic Acids administration & dosage, Bortezomib, Chromosome Deletion, Chromosomes, Human, Pair 17, Disease-Free Survival, Female, Follow-Up Studies, Humans, Immunoglobulin A blood, Interleukin-6 antagonists & inhibitors, Male, Melphalan administration & dosage, Middle Aged, Multiple Myeloma blood, Multiple Myeloma genetics, Prednisone administration & dosage, Pyrazines administration & dosage, Survival Rate, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Multiple Myeloma drug therapy, Multiple Myeloma mortality
- Abstract
Because interleukin-6 (IL-6) is considered important in the proliferation of early multiple myeloma (MM), we hypothesized that the addition of the anti-IL-6 monoclonal antibody siltuximab to the bortezomib-melphalan-prednisone (VMP) regimen would improve outcomes in transplant-ineligible patients with newly diagnosed MM. One hundred and six patients were randomized to receive 9 cycles of VMP or VMP plus siltuximab (11 mg/kg every 3 weeks) followed by siltuximab maintenance. Baseline characteristics were well balanced except for immunoglobulin A subtype and 17p deletions. With a complete response (CR) rate of 27% on siltuximab plus VMP (S+VMP) and 22% on VMP, the study did not confirm its hypothesis that the addition of siltuximab would increase the CR rate by at least 10%. Overall response rate was 88% on S+VMP and 80% on VMP, and at least very good partial response rates were 71% and 51% (P = .0382), respectively. Median progression-free survival (17 months) and 1-year overall survival (88%) were identical in the 2 arms. Grade ≥3 adverse-event incidence was 92% on S+VMP and 81% on VMP (P = .09), with trends toward more hematologic events and infections on S+VMP. Maintenance therapy with siltuximab was well tolerated. In conclusion, the addition of siltuximab to VMP did not improve the CR rate or long-term outcomes. This study was registered at http://clinicaltrials.gov as #NCT00911859., (© 2014 by The American Society of Hematology.)
- Published
- 2014
- Full Text
- View/download PDF
38. Osteoblastic niche supports the growth of quiescent multiple myeloma cells.
- Author
-
Chen Z, Orlowski RZ, Wang M, Kwak L, and McCarty N
- Subjects
- Animals, Cell Cycle Checkpoints, Cell Proliferation, Cell Tracking methods, Humans, Mice, Mice, Inbred NOD, Mice, SCID, Organic Chemicals analysis, Multiple Myeloma pathology, Neoplastic Stem Cells pathology, Osteoblasts pathology, Stem Cell Niche
- Abstract
The heterogeneity of multiple myeloma (MM) contributes to variable responses to therapy. In this study, we aim to correlate the heterogeneity of MM to the presence of quiescent cells using the PKH26 dye. We tracked the rare quiescent cells in different niches of the bone marrow by allowing the cells to cycle in vivo. Surprisingly, quiescent PKH(+) MM cells prefer to reside within the osteoblastic niches of the bone marrow (PKH(+)/OS) rather than the vascular (VS) niches or the spleen. These cells (PKH(+)/OS) displayed enhanced stemlike properties by forming colonies in semisolid medium. PKH(+) cells were highly tumorigenic compared with PKH(-) cells and were resistant to a variety of drugs. However, the levels of drug resistance were somewhat similar regardless of where the PKH(+) cells were isolated. Our data indicate that osteoblastic niches support the growth of quiescent PKH(+) cells and allow them to have stemlike functions.
- Published
- 2014
- Full Text
- View/download PDF
39. Mutation of NRAS but not KRAS significantly reduces myeloma sensitivity to single-agent bortezomib therapy.
- Author
-
Mulligan G, Lichter DI, Di Bacco A, Blakemore SJ, Berger A, Koenig E, Bernard H, Trepicchio W, Li B, Neuwirth R, Chattopadhyay N, Bolen JB, Dorner AJ, van de Velde H, Ricci D, Jagannath S, Berenson JR, Richardson PG, Stadtmauer EA, Orlowski RZ, Lonial S, Anderson KC, Sonneveld P, San Miguel JF, Esseltine DL, and Schu M
- Subjects
- Bortezomib, Cohort Studies, Dose-Response Relationship, Drug, Humans, Multiple Myeloma diagnosis, Multiple Myeloma genetics, Multiple Myeloma pathology, Prognosis, Proto-Oncogene Proteins p21(ras), Survival Analysis, Antineoplastic Agents therapeutic use, Boronic Acids therapeutic use, GTP Phosphohydrolases genetics, Membrane Proteins genetics, Multiple Myeloma drug therapy, Mutation, Proto-Oncogene Proteins genetics, Pyrazines therapeutic use, ras Proteins genetics
- Abstract
Various translocations and mutations have been identified in myeloma, and certain aberrations, such as t(4;14) and del17, are linked with disease prognosis. To investigate mutational prevalence in myeloma and associations between mutations and patient outcomes, we tested a panel of 41 known oncogenes and tumor suppressor genes in tumor samples from 133 relapsed myeloma patients participating in phase 2 or 3 clinical trials of bortezomib. DNA mutations were identified in 14 genes. BRAF as well as RAS genes were mutated in a large proportion of cases (45.9%) and these mutations were mutually exclusive. New recurrent mutations were also identified, including in the PDGFRA and JAK3 genes. NRAS mutations were associated with a significantly lower response rate to single-agent bortezomib (7% vs 53% in patients with mutant vs wild-type NRAS, P = .00116, Bonferroni-corrected P = .016), as well as shorter time to progression in bortezomib-treated patients (P = .0058, Bonferroni-corrected P = .012). However, NRAS mutation did not impact outcome in patients treated with high-dose dexamethasone. KRAS mutation did not reduce sensitivity to bortezomib or dexamethasone. These findings identify a significant clinical impact of NRAS mutation in myeloma and demonstrate a clear example of functional differences between the KRAS and NRAS oncogenes.
- Published
- 2014
- Full Text
- View/download PDF
40. Clinical, genomic, and imaging predictors of myeloma progression from asymptomatic monoclonal gammopathies (SWOG S0120).
- Author
-
Dhodapkar MV, Sexton R, Waheed S, Usmani S, Papanikolaou X, Nair B, Petty N, Shaughnessy JD Jr, Hoering A, Crowley J, Orlowski RZ, and Barlogie B
- Subjects
- Aged, Cohort Studies, Disease Progression, Female, Humans, Immunoglobulin Light Chains blood, Magnetic Resonance Imaging, Male, Middle Aged, Monoclonal Gammopathy of Undetermined Significance therapy, Multiple Myeloma therapy, Precancerous Conditions diagnosis, Precancerous Conditions genetics, Precancerous Conditions therapy, Proportional Hazards Models, Prospective Studies, Risk, Syndecan-1 metabolism, Time Factors, Monoclonal Gammopathy of Undetermined Significance diagnosis, Monoclonal Gammopathy of Undetermined Significance genetics, Multiple Myeloma diagnosis, Multiple Myeloma genetics
- Abstract
All cases of clinical myeloma (CMM) are preceded by an asymptomatic monoclonal gammopathy (AMG), classified as either monoclonal gammopathy of undetermined significance (MGUS) or asymptomatic multiple myeloma (AMM). We analyzed data from AMG patients (n = 331) enrolled in a prospective, observational clinical trial (S0120). Baseline data from clinical variables, gene expression profiles (GEP) of purified tumor cells, and findings of magnetic resonance imaging (MRI) were correlated with the risk of progression to CMM requiring therapy. GEP of purified tumor cells revealed that all molecular subtypes of CMM are also represented in the AMG phase. An increased risk score (>-0.26) (based on a 70-gene signature, GEP70) was an independent predictor of the risk of progression to CMM. Combination of elevated serum free light chain, M-spike, and GEP70 risk score identified a subset with high risk (67% at 2 years) of progression to CMM requiring therapy. Importantly, absence of these factors in AMM patients predicted low risk similar to MGUS. Detection of multiple (>1) focal lesions by MRI also conferred an increased risk of progression. These data demonstrate that signatures associated with high-risk CMM impact disease risk and support inclusion of genomic analysis in the clinical management of AMGs.
- Published
- 2014
- Full Text
- View/download PDF
41. Phase 2 dose-expansion study (PX-171-006) of carfilzomib, lenalidomide, and low-dose dexamethasone in relapsed or progressive multiple myeloma.
- Author
-
Wang M, Martin T, Bensinger W, Alsina M, Siegel DS, Kavalerchik E, Huang M, Orlowski RZ, and Niesvizky R
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols adverse effects, Dexamethasone administration & dosage, Dexamethasone adverse effects, Diarrhea chemically induced, Disease Progression, Disease-Free Survival, Dose-Response Relationship, Drug, Drug Administration Schedule, Fatigue chemically induced, Female, Humans, Kaplan-Meier Estimate, Lenalidomide, Lymphopenia chemically induced, Male, Middle Aged, Multiple Myeloma pathology, Neoplasm Recurrence, Local, Neutropenia chemically induced, Oligopeptides administration & dosage, Oligopeptides adverse effects, Remission Induction, Thalidomide administration & dosage, Thalidomide adverse effects, Thalidomide analogs & derivatives, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Multiple Myeloma drug therapy
- Abstract
We previously reported a phase 1b dose-escalation study of carfilzomib, lenalidomide, and low-dose dexamethasone (CRd) in relapsed or progressive multiple myeloma where the maximum planned dose (MPD) was carfilzomib 20 mg/m2 days 1 and 2 of cycle 1 and 27 mg/m2 days 8, 9, 15, 16, and thereafter; lenalidomide 25 mg days 1 to 21; and dexamethasone 40 mg once weekly on 28-day cycles. Herein, we present results from the phase 2 dose expansion at the MPD, focusing on the 52 patients enrolled in the MPD cohort. Median follow-up was 24.4 months. In the MPD cohort, overall response rate (ORR) was 76.9% with median time to response of 0.95 month (range, 0.5-4.6) and duration of response (DOR) of 22.1 months. Median progression-free survival was 15.4 months. ORR was 69.2% in bortezomib-refractory patients and 69.6% in lenalidomide-refractory patients with median DOR of 22.1 and 10.8 months, respectively. A median of 9.5 (range, 1-45) carfilzomib cycles were started with 7.7% of patients requiring carfilzomib dose reductions and 19.2% discontinuing CRd due to adverse events (AEs). Grade 3/4 AEs included lymphopenia (48.1%), neutropenia (32.7%), thrombocytopenia (19.2%), and anemia (19.2%). CRd at the MPD was well tolerated with robust, rapid, and durable responses.
- Published
- 2013
- Full Text
- View/download PDF
42. Biological effects of the Pim kinase inhibitor, SGI-1776, in multiple myeloma.
- Author
-
Cervantes-Gomez F, Chen LS, Orlowski RZ, and Gandhi V
- Subjects
- Animals, Autophagy drug effects, Cell Death drug effects, Cell Growth Processes drug effects, Cell Line, Tumor, Humans, Mice, Multiple Myeloma enzymology, Multiple Myeloma pathology, Proto-Oncogene Proteins c-pim-1 metabolism, Imidazoles pharmacology, Multiple Myeloma drug therapy, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-pim-1 antagonists & inhibitors, Pyridazines pharmacology
- Abstract
Background: Pim kinases are constitutively active serine/threonine/tyrosine kinases that are overexpressed in hematological malignancies such as multiple myeloma. Pim kinase substrates are involved in transcription, protein translation, cell proliferation, and apoptosis. SGI-1776 is a potent Pim kinase inhibitor that has proven to be cytotoxic to leukemia and lymphoma cells. Based on this background, we hypothesized that SGI-1776 treatment would result in myeloma cytotoxicity., Materials and Methods: To test this, myeloma cell lines and primary CD138(+) cells from myeloma patients were treated with SGI-1776 in a dose- and time-dependent manner, and effect on cell death and proliferation, induction of autophagy, and changes in cell cycle profile were measured., Results: SGI-1776 treatment resulted in limited apoptosis in cell lines (mean 30%) and CD138(+) cells (< 10%) assessed using Annexin-V/propidium iodide. Limited effect was observed in cell cycle profile or growth in cell lines. However, DNA synthesis was decreased by 70% at 3 μM (all time points) in U266 though this was not observed in MM.1S. In accordance, immunoblot analyses revealed no change in transcription (c-Myc and H3), or apoptotic (Bad) proteins that are substrates of Pim kinases. In contrast, autophagy, assessed using acridine orange staining, was induced with SGI-1776 treatment in both cell lines (U266, 25%-70%; MM.1S, 8%-52%) and CD138(+) cells (19%-21%). Immunoblot analyses of the autophagy LC3b marker and translation initiation proteins (phospho-p70S6K and 4E-BP1) corroborated autophagy induction., Conclusion: These data indicate that SGI-1776 treatment in myeloma cell lines and CD138(+) myeloma cells elicits its deleterious effects through inhibition of translation and induction of autophagy., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
43. An unusual case of aggressive systemic mastocytosis with associated refractory plasma cell myeloma.
- Author
-
Jain P, Verstovsek S, Orlowski RZ, Yap E, and Amin HM
- Subjects
- Female, Humans, Middle Aged, Mastocytosis, Systemic pathology, Multiple Myeloma pathology
- Published
- 2012
- Full Text
- View/download PDF
44. Targeting the insulin-like growth factor-1 receptor to overcome bortezomib resistance in preclinical models of multiple myeloma.
- Author
-
Kuhn DJ, Berkova Z, Jones RJ, Woessner R, Bjorklund CC, Ma W, Davis RE, Lin P, Wang H, Madden TL, Wei C, Baladandayuthapani V, Wang M, Thomas SK, Shah JJ, Weber DM, and Orlowski RZ
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols, Apoptosis drug effects, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Blotting, Western, Bortezomib, Cell Proliferation drug effects, Drug Synergism, Enzyme-Linked Immunosorbent Assay, Flow Cytometry, Gene Expression Profiling, Humans, Insulin-Like Growth Factor I metabolism, Mice, Mice, SCID, Multiple Myeloma metabolism, Multiple Myeloma mortality, Oligonucleotide Array Sequence Analysis, Receptor, IGF Type 1 metabolism, Survival Rate, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Boronic Acids pharmacology, Drug Resistance, Neoplasm, Imidazoles pharmacology, Multiple Myeloma drug therapy, Pyrazines pharmacology, Receptor, IGF Type 1 antagonists & inhibitors
- Abstract
Proteasome inhibition with bortezomib is a validated approach to the treatment of multiple myeloma, but drug resistance often emerges and limits its utility in the retreatment setting. To begin to identify some of the mechanisms involved, we developed bortezomib-resistant myeloma cell lines that, unlike previously reported models, showed no β5 subunit mutations. Instead, up-regulation of the insulin-like growth factor (IGF)-1 axis was identified, with increased autocrine and paracrine secretion of IGF-1, leading to increased activation of the IGF-1 receptor (IGF-1R). Exogenous IGF-1 reduced cellular sensitivity to bortezomib, whereas pharmacologic or small hairpin RNA-mediated IGF-1R suppression enhanced bortezomib sensitivity in cell lines and patient samples. In vitro studies with OSI-906, a clinically relevant dual IGF-1R and insulin receptor inhibitor, showed it acted synergistically with bortezomib, and potently resensitized bortezomib-resistant cell lines and patient samples to bortezomib. Importantly, OSI-906 in combination with bortezomib also overcame bortezomib resistance in an in vivo model of myeloma. Taken together, these data support the hypothesis that signaling through the IGF-1/IGF-1R axis contributes to acquired bortezomib resistance, and provide a rationale for combining bortezomib with IGF-1R inhibitors like OSI-906 to overcome or possibly prevent the emergence of bortezomib-refractory disease in the clinic.
- Published
- 2012
- Full Text
- View/download PDF
45. A phase 2 study of single-agent carfilzomib (PX-171-003-A1) in patients with relapsed and refractory multiple myeloma.
- Author
-
Siegel DS, Martin T, Wang M, Vij R, Jakubowiak AJ, Lonial S, Trudel S, Kukreti V, Bahlis N, Alsina M, Chanan-Khan A, Buadi F, Reu FJ, Somlo G, Zonder J, Song K, Stewart AK, Stadtmauer E, Kunkel L, Wear S, Wong AF, Orlowski RZ, and Jagannath S
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Humans, Male, Middle Aged, Multiple Myeloma mortality, Neoplasm Recurrence, Local mortality, Neoplasm Staging, Pilot Projects, Prognosis, Survival Rate, Drug Resistance, Neoplasm, Multiple Myeloma drug therapy, Neoplasm Recurrence, Local drug therapy, Oligopeptides therapeutic use, Salvage Therapy
- Abstract
Carfilzomib is a next-generation, selective proteasome inhibitor being evaluated for the treatment of relapsed and refractory multiple myeloma. In this open-label, single-arm phase 2 study (PX-171-003-A1), patients received single-agent carfilzomib 20 mg/m(2) intravenously twice weekly for 3 of 4 weeks in cycle 1, then 27 mg/m(2) for ≤ 12 cycles. The primary endpoint was overall response rate (≥ partial response). Secondary endpoints included clinical benefit response rate (≥ minimal response), duration of response, progression-free survival, overall survival, and safety. A total of 266 patients were evaluable for safety, 257 for efficacy; 95% were refractory to their last therapy; 80% were refractory or intolerant to both bortezomib and lenalidomide. Patients had median of 5 prior lines of therapy, including bortezomib, lenalidomide, and thalidomide. Overall response rate was 23.7% with median duration of response of 7.8 months. Median overall survival was 15.6 months. Adverse events (AEs) were manageable without cumulative toxicities. Common AEs were fatigue (49%), anemia (46%), nausea (45%), and thrombocytopenia (39%). Thirty-three patients (12.4%) experienced peripheral neuropathy, primarily grades 1 or 2. Thirty-three patients (12.4%) withdrew because of an AE. Durable responses and an acceptable tolerability profile in this heavily pretreated population demonstrate the potential of carfilzomib to offer meaningful clinical benefit. This trial was registered at www.clinicaltrials.gov as #NCT00511238.
- Published
- 2012
- Full Text
- View/download PDF
46. An open-label single-arm pilot phase II study (PX-171-003-A0) of low-dose, single-agent carfilzomib in patients with relapsed and refractory multiple myeloma.
- Author
-
Jagannath S, Vij R, Stewart AK, Trudel S, Jakubowiak AJ, Reiman T, Somlo G, Bahlis N, Lonial S, Kunkel LA, Wong A, Orlowski RZ, and Siegel DS
- Subjects
- Adult, Aged, Aged, 80 and over, Boronic Acids pharmacology, Bortezomib, Dose-Response Relationship, Drug, Drug Resistance, Neoplasm, Drug Tolerance, Female, Humans, Male, Middle Aged, Oligopeptides adverse effects, Pilot Projects, Proteasome Inhibitors adverse effects, Pyrazines pharmacology, Multiple Myeloma drug therapy, Oligopeptides administration & dosage, Proteasome Inhibitors administration & dosage, Secondary Prevention
- Abstract
Unlabelled: An open-label single-arm multicenter pilot phase II study of the next-generation selective proteasome inhibitor carfilzomib was conducted in 46 patients with relapsed and refractory multiple myeloma (MM) after ≥ 2 previous therapies. The best overall response rate (ORR) was 16.7%, with a median duration of response of 7.2 months. This pilot study was the first phase II single-agent trial conducted with carfilzomib., Background: Carfilzomib is a next-generation selective proteasome inhibitor that irreversibly binds its target and has demonstrated single-agent activity in patients with bortezomib-resistant multiple myeloma (MM). PX-171-003-A0, an open-label single-arm multicenter pilot phase II study, enrolled 46 patients with relapsed MM after ≥ 2 previous therapies including bortezomib and an immunomodulator (thalidomide or lenalidomide) and disease refractory to the last treatment regimen preceding study entry., Methods: Patients received carfilzomib 20 mg/m(2) intravenously on days 1, 2, 8, 9, 15, and 16 every 28 days for up to 12 cycles. Responses in 42 evaluable patients were assessed per International Myeloma Working Group Uniform Response Criteria, with minimal response assessed per European Group for Blood and Marrow Transplantation (EBMT) criteria., Results: The primary endpoint of best ORR was 16.7%, including 7 partial responses. Median duration of response was 7.2 months. Clinical benefit response (CBR) rate was 23.8% with a median duration of response of 13.8 months. The most common treatment-emergent adverse events (AEs) of any grade were anemia (73.9%), fatigue (69.6%), and thrombocytopenia (50.0%). Notably, peripheral neuropathy and neuropathy-related AEs were generally mild and infrequent., Conclusion: This pilot study was the first phase II single-agent trial conducted with carfilzomib. Based on these findings, the study was amended to test a higher carfilzomib dose in an additional 250 patients (PX-171-003-A1)., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
47. Proteasome inhibitors in multiple myeloma: 10 years later.
- Author
-
Moreau P, Richardson PG, Cavo M, Orlowski RZ, San Miguel JF, Palumbo A, and Harousseau JL
- Subjects
- Animals, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Boronic Acids adverse effects, Boronic Acids pharmacology, Boronic Acids therapeutic use, Bortezomib, Cysteine Proteinase Inhibitors adverse effects, Cysteine Proteinase Inhibitors pharmacology, Humans, Medical Oncology methods, Pyrazines adverse effects, Pyrazines pharmacology, Pyrazines therapeutic use, Time Factors, Cysteine Proteinase Inhibitors therapeutic use, Medical Oncology trends, Multiple Myeloma drug therapy, Proteasome Inhibitors
- Abstract
Proteasome inhibition has emerged as an important therapeutic strategy in multiple myeloma (MM). Since the publication of the first phase 1 trials of bortezomib 10 years ago, this first-in-class proteasome inhibitor (PI) has contributed substantially to the observed improvement in survival in MM patients over the past decade. Although first approved as a single agent in the relapsed setting, bortezomib is now predominantly used in combination regimens. Furthermore, the standard twice-weekly schedule may be replaced by weekly infusion, especially when bortezomib is used as part of combination regimens in frontline therapy. Indeed, bortezomib is an established component of induction therapy for patients eligible or ineligible for autologous stem cell transplantation. Bortezomib has also been incorporated into conditioning regimens before autologous stem cell transplantation, as well as into post-ASCT consolidation therapy, and in the maintenance setting. In addition, a new route of bortezomib administration, subcutaneous infusion, has recently been approved. Recently, several new agents have been introduced into the clinic, including carfilzomib, marizomib, and MLN9708, and trials investigating these "second-generation" PIs in patients with relapsed/refractory MMs have demonstrated positive results. This review provides an overview of the role of PIs in the treatment of MM, focusing on developments over the past decade.
- Published
- 2012
- Full Text
- View/download PDF
48. The zinc finger transcription factor ZKSCAN3 promotes prostate cancer cell migration.
- Author
-
Zhang X, Jing Y, Qin Y, Hunsucker S, Meng H, Sui J, Jiang Y, Gao L, An G, Yang N, Orlowski RZ, and Yang L
- Subjects
- Animals, Cell Line, Tumor, Cells, Cultured, Gene Knockdown Techniques, Humans, Immunohistochemistry, Male, Mice, Prostatic Neoplasms pathology, Transcription Factors deficiency, Transcription Factors metabolism, Transfection, Cell Movement genetics, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism, Transcription Factors genetics, Zinc Fingers genetics
- Abstract
In our previous studies, ZKSCAN3 was demonstrated to be over-expressed in invasive colonic tumor cells and their liver metastases, but minimally expressed in adjacent non-transformed tissues. Further preliminary data showed that ZKSCAN3 was expressed in a majority of prostate cancer patient samples, but not in normal prostate tissues. Moreover, the ZKSCAN3 protein is highly expressed in the PC3 prostate cancer cell line, which has high metastatic potential, but little expression was observed in non-metastatic prostate cancer cell lines. Thus, we hypothesized that ZKSCAN3 could participate in tumor metastasis by regulating tumor cell migration. To test this hypothesis, ZKSCAN3 mRNA was knocked down by ZKSCAN3 specific shRNA in PC3 cells and a significant decrease in cell motility was observed. In contrast, when ZKSCAN3 cDNA was overexpressed in PC3 cells, cell detachment was observed and suspension culture induced apoptosis was greatly decreased, suggesting that ZKSCAN3 is able to enhance PC3 cell survival under anoikis stress. Additional wound healing and invasion assays showed that cell migration was enhanced by ZKSCAN3 expression. Interestingly, the ZKSCAN3 gene was amplified in 26% (5/19) of metastatic prostate cancers and 20% (1/5) of lymph node metastases, but there was no amplification found in primary prostate cancers, further supporting the role of ZKSCAN3 in tumor cell migration. In vivo studies using orthotopic tumor models indicated that overexpression of ZKSCAN3 significantly enhanced tumorigenicity. Taken together, we provide evidence that ZKSCAN3, a zinc finger transcription factor, plays a critical role in promoting prostate cancer cell migration., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
49. An open-label, single-arm, phase 2 (PX-171-004) study of single-agent carfilzomib in bortezomib-naive patients with relapsed and/or refractory multiple myeloma.
- Author
-
Vij R, Wang M, Kaufman JL, Lonial S, Jakubowiak AJ, Stewart AK, Kukreti V, Jagannath S, McDonagh KT, Alsina M, Bahlis NJ, Reu FJ, Gabrail NY, Belch A, Matous JV, Lee P, Rosen P, Sebag M, Vesole DH, Kunkel LA, Wear SM, Wong AF, Orlowski RZ, and Siegel DS
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Agents administration & dosage, Antineoplastic Agents adverse effects, Bortezomib, Cohort Studies, Demography, Disease-Free Survival, Female, Humans, Male, Middle Aged, Oligopeptides administration & dosage, Oligopeptides adverse effects, Recurrence, Time Factors, Treatment Outcome, Antineoplastic Agents therapeutic use, Boronic Acids therapeutic use, Multiple Myeloma drug therapy, Oligopeptides therapeutic use, Pyrazines therapeutic use
- Abstract
Carfilzomib is a selective proteasome inhibitor that binds irreversibly to its target. In phase 1 studies, carfilzomib elicited promising responses and an acceptable toxicity profile in patients with relapsed and/or refractory multiple myeloma (R/R MM). In the present phase 2, multicenter, open-label study, 129 bortezomib-naive patients with R/R MM (median of 2 prior therapies) were separated into Cohort 1, scheduled to receive intravenous carfilzomib 20 mg/m(2) for all treatment cycles, and Cohort 2, scheduled to receive 20 mg/m(2) for cycle 1 and then 27 mg/m(2) for all subsequent cycles. The primary end point was an overall response rate (≥ partial response) of 42.4% in Cohort 1 and 52.2% in Cohort 2. The clinical benefit response (overall response rate + minimal response) was 59.3% and 64.2% in Cohorts 1 and 2, respectively. Median duration of response was 13.1 months and not reached, and median time to progression was 8.3 months and not reached, respectively. The most common treatment-emergent adverse events were fatigue (62.0%) and nausea (48.8%). Single-agent carfilzomib elicited a low incidence of peripheral neuropathy-17.1% overall (1 grade 3; no grade 4)-in these pretreated bortezomib-naive patients. The results of the present study support the use of carfilzomib in R/R MM patients. This trial is registered at www.clinicaltrials.gov as NCT00530816.
- Published
- 2012
- Full Text
- View/download PDF
50. Dysfunction of the TP53 tumor suppressor gene in lymphoid malignancies.
- Author
-
Xu-Monette ZY, Medeiros LJ, Li Y, Orlowski RZ, Andreeff M, Bueso-Ramos CE, Greiner TC, McDonnell TJ, and Young KH
- Subjects
- Humans, Lymphocytes metabolism, Lymphoma metabolism, MicroRNAs genetics, Transcription, Genetic immunology, Tumor Suppressor Protein p53 metabolism, Lymphocytes immunology, Lymphoma genetics, Lymphoma immunology, Signal Transduction immunology, Tumor Suppressor Protein p53 genetics
- Abstract
Mutations of the TP53 gene and dysregulation of the TP53 pathway are important in the pathogenesis of many human cancers, including lymphomas. Tumor suppression by p53 occurs via both transcription-dependent activities in the nucleus by which p53 regulates transcription of genes involved in cell cycle, DNA repair, apoptosis, signaling, transcription, and metabolism; and transcription-independent activities that induces apoptosis and autophagy in the cytoplasm. In lymphoid malignancies, the frequency of TP53 deletions and mutations is lower than in other types of cancer. Nonetheless, the status of TP53 is an independent prognostic factor in most lymphoma types. Dysfunction of TP53 with wild-type coding sequence can result from deregulated gene expression, stability, and activity of p53. To overcome TP53 pathway inactivation, therapeutic delivery of wild-type p53, activation of mutant p53, inhibition of MDM2-mediated degradation of p53, and activation of p53-dependent and -independent apoptotic pathways have been explored experimentally and in clinical trials. We review the mechanisms of TP53 dysfunction, recent advances implicated in lymphomagenesis, and therapeutic approaches to overcoming p53 inactivation.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.