Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Raymond L. Comenzo is active.

Publication


Featured researches published by Raymond L. Comenzo.


Leukemia | 2009

International Myeloma Working Group guidelines for serum-free light chain analysis in multiple myeloma and related disorders

Angela Dispenzieri; Robert A. Kyle; Giampaolo Merlini; Jesús F. San Miguel; H. Ludwig; Roman Hájek; A. Palumbo; Sundar Jagannath; J. Bladé; Sagar Lonial; M. Dimopoulos; Raymond L. Comenzo; Hermann Einsele; Bart Barlogie; Kenneth C. Anderson; Morie A. Gertz; Jean Luc Harousseau; Michel Attal; Patrizia Tosi; Pieter Sonneveld; Mario Boccadoro; Gareth J. Morgan; Paul G. Richardson; Orhan Sezer; M.V. Mateos; Michele Cavo; Doug Joshua; Ingemar Turesson; Wenming Chen; Kazuyuki Shimizu

The serum immunoglobulin-free light chain (FLC) assay measures levels of free κ and λ immunoglobulin light chains. There are three major indications for the FLC assay in the evaluation and management of multiple myeloma and related plasma cell disorders (PCD). In the context of screening, the serum FLC assay in combination with serum protein electrophoresis (PEL) and immunofixation yields high sensitivity, and negates the need for 24-h urine studies for diagnoses other than light chain amyloidosis (AL). Second, the baseline FLC measurement is of major prognostic value in virtually every PCD. Third, the FLC assay allows for quantitative monitoring of patients with oligosecretory PCD, including AL, oligosecretory myeloma and nearly two-thirds of patients who had previously been deemed to have non-secretory myeloma. In AL patients, serial FLC measurements outperform PEL and immunofixation. In oligosecretory myeloma patients, although not formally validated, serial FLC measurements reduce the need for frequent bone marrow biopsies. In contrast, there are no data to support using FLC assay in place of 24-h urine PEL for monitoring or for serial measurements in PCD with measurable disease by serum or urine PEL. This paper provides consensus guidelines for the use of this important assay, in the diagnosis and management of clonal PCD.


Leukemia | 2009

International myeloma working group consensus statement and guidelines regarding the current role of imaging techniques in the diagnosis and monitoring of multiple Myeloma

Meletios A. Dimopoulos; E Terpos; Raymond L. Comenzo; Patrizia Tosi; Meral Beksac; Orhan Sezer; David Siegel; Henk M. Lokhorst; Shaji Kumar; S V Rajkumar; Ruben Niesvizky; L A Moulopoulos; Brian G. M. Durie

Several imaging technologies are used for the diagnosis and management of patients with multiple myeloma (MM). Conventional radiography, computed tomography (CT), magnetic resonance imaging (MRI) and nuclear medicine imaging are all used in an attempt to better clarify the extent of bone disease and soft tissue disease in MM. This review summarizes all available data in the literature and provides recommendations for the use of each of the technologies. Conventional radiography still remains the ‘gold standard’ of the staging procedure of newly diagnosed and relapsed myeloma patients. MRI gives information complementary to skeletal survey and is recommended in MM patients with normal conventional radiography and in all patients with an apparently solitary plasmacytoma of bone. Urgent MRI or CT (if MRI is not available) is the diagnostic procedure of choice to assess suspected cord compression. Bone scintigraphy has no place in the routine staging of myeloma, whereas sequential dual-energy X-ray absorptiometry scans are not recommended. Positron emission tomography/CT or MIBI imaging are also not recommended for routine use in the management of myeloma patients, although both techniques may be useful in selected cases that warrant clarification of previous imaging findings, but such an approach should ideally be made within the context of a clinical trial.


Journal of Clinical Oncology | 2012

New Criteria for Response to Treatment in Immunoglobulin Light Chain Amyloidosis Based on Free Light Chain Measurement and Cardiac Biomarkers: Impact on Survival Outcomes

Giovanni Palladini; Angela Dispenzieri; Morie A. Gertz; Shaji Kumar; Ashutosh D. Wechalekar; Philip N. Hawkins; Stefan Schönland; Ute Hegenbart; Raymond L. Comenzo; Efstathios Kastritis; Meletios A. Dimopoulos; Arnaud Jaccard; Catherine Klersy; Giampaolo Merlini

PURPOSE To identify the criteria for hematologic and cardiac response to treatment in immunoglobulin light chain (AL) amyloidosis based on survival analysis of a large patient population. PATIENTS AND METHODS We gathered for analysis 816 patients with AL amyloidosis from seven referral centers in the European Union and the United States. A different cohort of 374 patients prospectively evaluated at the Pavia Amyloidosis Research and Treatment Center was used for validation. Data was available for all patients before and 3 and/or 6 months after initiation of first-line therapy. The prognostic relevance of different criteria for hematologic and cardiac response was assessed. RESULTS There was a strong correlation between the extent of reduction of amyloidogenic free light chains (FLCs) and improvement in survival. This allowed the identification of four levels of response: amyloid complete response (normal FLC ratio and negative serum and urine immunofixation), very good partial response (difference between involved and uninvolved FLCs [dFLC] < 40 mg/L), partial response (dFLC decrease > 50%), and no response. Cardiac involvement is the major determinant of survival, and changes in cardiac function after therapy can be reliably assessed using the cardiac biomarker N-terminal natriuretic peptide type B (NT-proBNP). Changes in FLC and NT-proBNP predicted survival as early as 3 months after treatment initiation. CONCLUSION This study identifies and validates new criteria for response to first-line treatment in AL amyloidosis, based on their association with survival in large patient populations, and offers surrogate end points for clinical trials.


Journal of Clinical Oncology | 2010

Renal Impairment in Patients With Multiple Myeloma: A Consensus Statement on Behalf of the International Myeloma Working Group

Meletios A. Dimopoulos; Evangelos Terpos; Asher Chanan-Khan; Nelson Leung; Heinz Ludwig; Sundar Jagannath; Ruben Niesvizky; Sergio Giralt; Jean Paul Fermand; Joan Bladé; Raymond L. Comenzo; Orhan Sezer; Antonio Palumbo; Jean Luc Harousseau; Paul G. Richardson; Bart Barlogie; Kenneth C. Anderson; Pieter Sonneveld; Patrizia Tosi; Michele Cavo; S. Vincent Rajkumar; Brian G. M. Durie; Jesús F. San Miguel

Renal impairment is a common complication of multiple myeloma (MM). The estimated glomerular filtration rate (eGFR) using the Modification of Diet in Renal Disease formula is the recommended method for the assessment of renal function in patients with MM with stabilized serum creatinine. In acute renal injury, the RIFLE (risk, injury, failure, loss and end-stage kidney disease) and Acute Renal Injury Network criteria seem to be appropriate to define the severity of renal impairment. Novel criteria based on eGFR measurements are recommended for the definition of the reversibility of renal impairment. Rapid intervention to reverse renal dysfunction is critical for the management of these patients, especially for those with light chain cast nephropathy. Bortezomib with high-dose dexamethasone is considered as the treatment of choice for such patients. There is limited experience with thalidomide in patients with myeloma with renal impairment. Thus, thalidomide can be carefully administered, mainly in the context of well-designed clinical trials, to evaluate if it can improve the rapidity and probability of response that is produced by the combination with bortezomib and high-dose dexamethasone. Lenalidomide is effective in this setting and can reverse renal insufficiency in a significant subset of patients, when it is given at reduced doses, according to renal function. The role of plasma exchange in patients with suspected light chain cast nephropathy and renal impairment is controversial. High-dose melphalan (140 mg/m(2)) and autologous stem-cell transplantation should be limited to younger patients with chemosensitive disease.


The New England Journal of Medicine | 2001

Deep Venous Thrombosis and Thalidomide Therapy for Multiple Myeloma

Keren Osman; Raymond L. Comenzo; S. Vincent Rajkumar

To the Editor: Thalidomide is effective for the treatment of refractory myeloma.1 Its role in the initial treatment of the disease is under active investigation.2,3 We report the occurrence of deep...


Blood | 2009

Mobilization in myeloma revisited: IMWG consensus perspectives on stem cell collection following initial therapy with thalidomide, lenalidomide or bortezomib- containing regimens

Shaji Kumar; Sergio Giralt; Edward A. Stadtmauer; Jean Luc Harousseau; Antonio Palumbo; William Bensinger; Raymond L. Comenzo; Suzanne Lentzsch; Nikhil C. Munshi; Ruben Niesvizky; Jesús F. San Miguel; Heinz Ludwig; Leif Bergsagel; Joan Bladé; Sagar Lonial; Kenneth C. Anderson; Patrizia Tosi; Pieter Sonneveld; Orhan Sezer; David H. Vesole; Michele Cavo; Hermann Einsele; Paul G. Richardson; Brian G. M. Durie; S. Vincent Rajkumar

The past decade has witnessed a paradigm shift in the initial treatment of multiple myeloma with the introduction of novel agents such as thalidomide, lenalidomide, and bortezomib, leading to improved outcomes. High-dose therapy and autologous stem cell transplantation remains an important therapeutic option for patients with multiple myeloma eligible for the procedure. Before the advent of the novel agents, patients underwent stem cell collection prior to significant alkylating agent exposure, given its potential deleterious effect on stem cell collection. With increasing use of the novel agents in the upfront setting, several reports have emerged raising concerns about their impact on the ability to collect stem cells. An expert panel of the International Myeloma Working Group (IMWG) was convened to examine the implications of these therapies on stem collection in patients with myeloma and to develop recommendations for addressing these issues. Here we summarize the currently available data and present our perspective on the problem and potential options to overcome this problem. Specifically, we recommend early mobilization of stem cells, preferably within the first 4 cycles of initial therapy, in patients treated with novel agents and encourage participation in clinical trials evaluating novel approaches to stem cell mobilization.


Lancet Oncology | 2011

Autologous haemopoietic stem-cell transplantation followed by allogeneic or autologous haemopoietic stem-cell transplantation in patients with multiple myeloma (BMT CTN 0102): a phase 3 biological assignment trial

Amrita Krishnan; Marcelo C. Pasquini; Brent R. Logan; Edward A. Stadtmauer; David H. Vesole; Edwin P. Alyea; Joseph H. Antin; Raymond L. Comenzo; Stacey Goodman; Parameswaran Hari; Ginna G. Laport; Muzaffar H. Qazilbash; Scott D. Rowley; Firoozeh Sahebi; George Somlo; Dan T. Vogl; Daniel J. Weisdorf; Marian Ewell; Juan Wu; Nancy L. Geller; Mary M. Horowitz; Sergio Giralt; David G. Maloney

BACKGROUND Autologous haemopoietic stem-cell transplantation (HSCT) improves survival in patients with multiple myeloma, but disease progression remains an issue. Allogeneic HSCT might reduce disease progression, but can be associated with high treatment-related mortality. Thus, we aimed to assess effectiveness of allogeneic HSCT with non-myeloablative conditioning after autologous HSCT compared with tandem autologous HSCT. METHODS In our phase 3 biological assignment trial, we enrolled patients with multiple myeloma attending 37 transplant centres in the USA. Patients (<70 years old) with adequate organ function who had completed at least three cycles of systemic antimyeloma therapy within the past 10 months were eligible for inclusion. We assigned patients to receive an autologous HSCT followed by an allogeneic HSCT (auto-allo group) or tandem autologous HSCTs (auto-auto group) on the basis of the availability of an HLA-matched sibling donor. Patients in the auto-auto group subsequently underwent a random allocation (1:1) to maintenance therapy (thalidomide plus dexamethasone) or observation. To avoid enrolment bias, we classified patients as standard risk or high risk on the basis of cytogenetics and β2-microglobulin concentrations. We used the Kaplan-Meier method to estimate differences in 3-year progression-free survival (PFS; primary endpoint) between patients with standard-risk disease in the auto-allo group and the best results from the auto-auto group (maintenance, observation, or pooled). This study is registered with ClinicalTrials.gov, number NCT00075829. FINDINGS Between Dec 17, 2003, and March 30, 2007, we enrolled 710 patients, of whom 625 had standard-risk disease and received an autologous HSCT. 156 (83%) of 189 patients with standard-risk disease in the auto-allo group and 366 (84%) of 436 in the auto-auto group received a second transplant. 219 patients in the auto-auto group were randomly assigned to observation and 217 to receive maintenance treatment, of whom 168 (77%) completed this treatment. PFS and overall survival did not differ between maintenance and observation groups and pooled data were used. Kaplan-Meier estimates of 3-year PFS were 43% (95% CI 36-51) in the auto-allo group and 46% (42-51) in the auto-auto group (p=0·671); overall survival also did not differ at 3 years (77% [95% CI 72-84] vs 80% [77-84]; p=0·191). Within 3 years, 87 (46%) of 189 patients in the auto-allo group had grade 3-5 adverse events as did 185 (42%) of 436 patients in the auto-auto group. The adverse events that differed most between groups were hyperbilirubinaemia (21 [11%] patients in the auto-allo group vs 14 [3%] in the auto-auto group) and peripheral neuropathy (11 [6%] in the auto-allo group vs 52 [12%] in the auto-auto group). INTERPRETATION Non-myeloablative allogeneic HSCT after autologous HSCT is not more effective than tandem autologous HSCT for patients with standard-risk multiple myeloma. Further enhancement of the graft versus myeloma effect and reduction in transplant-related mortality are needed to improve the allogeneic HSCT approach. FUNDING US National Heart, Lung, and Blood Institute and the National Cancer Institute.


Leukemia | 2009

International myeloma working group (IMWG) consensus statement and guidelines regarding the current status of stem cell collection and high-dose therapy for multiple myeloma and the role of plerixafor (AMD 3100).

Sergio Giralt; Edward A. Stadtmauer; Jean Luc Harousseau; A. Palumbo; William Bensinger; Raymond L. Comenzo; Shaji Kumar; Nikhil C. Munshi; Angela Dispenzieri; Robert A. Kyle; Giampaolo Merlini; J. F. San Miguel; H. Ludwig; Roman Hájek; Sundar Jagannath; J. Bladé; Sagar Lonial; M. A. Dimopoulos; Hermann Einsele; Bart Barlogie; Kenneth C. Anderson; Morie A. Gertz; Michel Attal; Patrizia Tosi; Pieter Sonneveld; Mario Boccadoro; Gareth J. Morgan; Orhan Sezer; M.V. Mateos; Michele Cavo

Multiple myeloma is the most common indication for high-dose chemotherapy with autologous stem cell support (ASCT) in North America today. Stem cell procurement for ASCT has most commonly been performed with stem cell mobilization using colony-stimulating factors with or without prior chemotherapy. The target CD34+ cell dose to be collected as well as the number of apheresis performed varies throughout the country, but a minimum of 2 million CD34+ cells/kg has been traditionally used for the support of one cycle of high-dose therapy. With the advent of plerixafor (AMD3100) (a novel stem cell mobilization agent), it is pertinent to review the current status of stem cell mobilization for myeloma as well as the role of autologous stem cell transplantation in this disease. On June 1, 2008, a panel of experts was convened by the International Myeloma Foundation to address issues regarding stem cell mobilization and autologous transplantation in myeloma in the context of new therapies. The panel was asked to discuss a variety of issues regarding stem cell collection and transplantation in myeloma especially with the arrival of plerixafor. Herein, is a summary of their deliberations and conclusions.


Leukemia | 2012

Management of treatment-emergent peripheral neuropathy in multiple myeloma

Paul G. Richardson; Michel Delforge; Meral Beksac; Patrick Y. Wen; J L Jongen; Orhan Sezer; Evangelos Terpos; Nikhil C. Munshi; A. Palumbo; S V Rajkumar; Jean Luc Harousseau; P. Moreau; Hervé Avet-Loiseau; Jae Hoon Lee; Michele Cavo; Giampaolo Merlini; Peter M. Voorhees; Wee Joo Chng; Amitabha Mazumder; Saad Z Usmani; Hermann Einsele; Raymond L. Comenzo; Robert Z. Orlowski; David H. Vesole; Juan José Lahuerta; Ruben Niesvizky; David Siegel; M.V. Mateos; M. A. Dimopoulos; Sagar Lonial

Peripheral neuropathy (PN) is one of the most important complications of multiple myeloma (MM) treatment. PN can be caused by MM itself, either by the effects of the monoclonal protein or in the form of radiculopathy from direct compression, and particularly by certain therapies, including bortezomib, thalidomide, vinca alkaloids and cisplatin. Clinical evaluation has shown that up to 20% of MM patients have PN at diagnosis and as many as 75% may experience treatment-emergent PN during therapy. The incidence, symptoms, reversibility, predisposing factors and etiology of treatment-emergent PN vary among MM therapies, with PN incidence also affected by the dose, schedule and combinations of potentially neurotoxic agents. Effective management of treatment-emergent PN is critical to minimize the incidence and severity of this complication, while maintaining therapeutic efficacy. Herein, the state of knowledge regarding treatment-emergent PN in MM patients and current management practices are outlined, and recommendations regarding optimal strategies for PN management during MM treatment are provided. These strategies include early and regular monitoring with neurological evaluation, with dose modification and treatment discontinuation as indicated. Areas requiring further research include the development of MM-specific, patient-focused assessment tools, pharmacogenomic analysis of patient DNA, and trials to assess the efficacy of pharmacological interventions.


Cancer Research | 2006

Pyridone 6, A Pan-Janus-Activated Kinase Inhibitor, Induces Growth Inhibition of Multiple Myeloma Cells

Laura Pedranzini; Tobias Dechow; Marjan Berishaj; Raymond L. Comenzo; Ping Zhou; Janeen Azare; William G. Bornmann; Jacqueline Bromberg

Interleukin-6 (IL-6) and the subsequent Janus-activated kinase (JAK)-dependent signaling pathways play a critical role in the pathogenesis of multiple myeloma. Here, we compared the sensitivity and specificity of a novel pan-JAK inhibitor, tetracyclic pyridone 6 (P6), with that of AG490 in a panel of myeloma-derived cell lines. P6 induced growth arrest and subsequent apoptosis of the IL-6-dependent hybridoma and myeloma-derived cell lines (B9 and INA-6) grown either in IL-6-containing medium or in the presence of bone marrow-derived stromal cells (BMSC) using much lower concentrations of drug and with significantly faster kinetics than AG490. Myeloma-derived cell lines, which either express constitutively activated JAK/signal transducers and activators of transcription (STAT) 3 (U266) or are IL-6 growth stimulated (KMS11), are partially growth inhibited by P6. However, P6 does not inhibit the growth of myeloma-derived cell lines lacking activated JAKs/STATs nor does it inhibit mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) kinase activity compared with AG490, which led to activation of ERK and induced robust apoptosis of all the examined cell lines. Finally, P6 inhibited the growth of primary myeloma patient samples grown in the presence of BMSCs. Thus, P6 is a more sensitive and specific inhibitor of JAK-STAT3 activity compared with AG490 and potently inhibited the growth of primary myeloma cells and myeloma-derived cell lines grown on BMSCs.

Collaboration


Dive into the Raymond L. Comenzo's collaboration.

Top Co-Authors

Avatar

Heather Landau

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hani Hassoun

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Adam D. Cohen

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Martin Fleisher

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge