1
|
Wang S, Mouliere F, Pegtel DM, Chamuleau MED. Turning the tide in aggressive lymphoma: liquid biopsy for risk-adapted treatment strategies. Trends Mol Med 2024; 30:660-672. [PMID: 38692937 DOI: 10.1016/j.molmed.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/30/2024] [Accepted: 04/04/2024] [Indexed: 05/03/2024]
Abstract
Diffuse large B cell lymphoma (DLBCL) exhibits significant biological and clinical heterogeneity that presents challenges for risk stratification and disease surveillance. Existing tools for risk stratification, including the international prognostic index (IPI), tissue molecular analyses, and imaging, have limited accuracy in predicting outcomes. The therapeutic landscape for aggressive lymphoma is rapidly evolving, and there is a pressing need to identify patients at risk of refractory or relapsed (R/R) disease in the context of personalized therapy. Liquid biopsy, a minimally invasive method for cancer signal detection, has been explored to address these challenges. We review advances in liquid biopsy strategies focusing on circulating nucleic acids in DLBCL patients and highlight their clinical potential. We also provide recommendations for biomarker-guided trials to support risk-adapted treatment modalities.
Collapse
Affiliation(s)
- Steven Wang
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands; Imaging and Biomarkers, Cancer Center Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands
| | - Florent Mouliere
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands; Imaging and Biomarkers, Cancer Center Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands; Cancer Research UK National Biomarker Centre, University of Manchester, Wilmslow Road, Manchester, UK
| | - D Michiel Pegtel
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands; Imaging and Biomarkers, Cancer Center Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands
| | - Martine E D Chamuleau
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands; Imaging and Biomarkers, Cancer Center Amsterdam, De Boelelaan, 1117, Amsterdam, The Netherlands.
| |
Collapse
|
2
|
Song X, Wei Y, Zhang N, Sun X, Kang L. A Case of Diffuse Large B Cell Lymphoma With Rapidly Developing Abdominal Distension As the Only First Clinical Presentation. Cureus 2024; 16:e64837. [PMID: 39161525 PMCID: PMC11332976 DOI: 10.7759/cureus.64837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2024] [Indexed: 08/21/2024] Open
Abstract
This article presents a case study of diffuse large B-cell lymphoma in an elderly patient whose initial clinical manifestation was rapidly developing abdominal distension. The article delves into the patient's diagnostic and treatment journey, highlights treatment insights, and reviews relevant literature. The aim is to enhance the clinical diagnosis accuracy for elderly lymphoma patients presenting with a singular atypical symptom, ultimately optimizing treatment plans and enriching clinicians' knowledge of the disease.
Collapse
Affiliation(s)
- Xiaoda Song
- Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, CHN
| | - Yuchen Wei
- Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, CHN
| | - Ning Zhang
- Department of Geriatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, CHN
| | - Xiaohong Sun
- Department of Geriatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, CHN
| | - Lin Kang
- Department of Geriatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, CHN
| |
Collapse
|
3
|
Rutherford SC, Yin J, Pederson LD, Blum KA, Martin P, Jung SH, Grant B, Rosenbaum C, Cheson BD, Bartlett NL, Mandrekar SJ, Leonard JP. Impact of imaging frequency on progression-free survival in Alliance trials enrolling patients with follicular lymphoma. Blood Adv 2024; 8:1464-1468. [PMID: 38266151 PMCID: PMC10955638 DOI: 10.1182/bloodadvances.2023012090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/10/2024] [Accepted: 01/14/2024] [Indexed: 01/26/2024] Open
Affiliation(s)
- Sarah C. Rutherford
- Division of Hematology and Medical Oncology, Weill Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY
| | - Jun Yin
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN
| | - Levi D. Pederson
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN
| | - Kristie A. Blum
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Peter Martin
- Division of Hematology and Medical Oncology, Weill Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY
| | - Sin-Ho Jung
- Alliance Statistics and Data Management Center, Biostatistics and Bioinformatics, Duke Cancer Institute, Duke University, Durham, NC
| | - Barbara Grant
- Division of Hematology and Oncology, University of Vermont, Burlington, VT
| | - Cara Rosenbaum
- Division of Hematology and Medical Oncology, Weill Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY
| | | | - Nancy L. Bartlett
- Division of Oncology, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO
| | | | - John P. Leonard
- Division of Hematology and Medical Oncology, Weill Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY
| |
Collapse
|
4
|
Hou Y, Zi J, Liu S, Ge Q, Ge Z. Mutational profiling of circulating tumor DNA and clinical characteristics in lymphoma: Based on next generation sequencing. Mol Carcinog 2023; 62:200-209. [PMID: 36300887 DOI: 10.1002/mc.23476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 07/10/2022] [Accepted: 09/30/2022] [Indexed: 01/21/2023]
Abstract
Liquid biopsy has been experimented with to identify the mutation of lymphoma based on next-generation sequencing (NGS). We applied NGS analysis to circulating tumor DNA (ctDNA) in 20 lymphoma patients. Then, we compared treatment outcomes, and clinical characteristics among these patients, then investigated mutational profiling. Two independent cohorts of 241 patients with mature B cell lymphoma in Mature B-cell malignancies data set (MBN) data set and 50 diffuse large B-cell lymphoma (DLBCL) patients in DLBCL data set, were used to examine the association between gene mutations and prognosis. We found ctDNA positive group had significantly more relapsed/PD (7/12, 58.3%) and less CR/PR patients (1/12, 8.3%) compared to negative group (0, 0%) (5/8, 62.5%) (p < 0.001). Somatic alterations were identified in 12 of 20 patients and the total 11 mutations were: Ataxia telangiectasia mutated (ATM), TP53, BCL2, BTG2, CD28, EP300, IDH2, IRF8, JAK3, NOTCH1, and NRAS. ATM (S2168L) was found in SLL and TLBL for the first time. BTG2 (c.292_293del), CD28 (P119T), IRF8 (E74D) and NOTCH1 (c.4348 G > A) were newly detected in DLBCL, angioimmunoblastic T-cell lymphoma, primary central nervous system lymphoma, and BCL for the first time respectively. We also disclosed an unreported mutation EP300 (c.1058_1059insC) in DLBCL. Our cases implied ctDNA detection consistent with the FISH of tissue samples to some extent, speculating new molecular subtypes of DLBCL, finding some potential drug-resistant mutations, and suggesting disease recurrence. Moreover, in MBN and DLBCL datasets, patients with TP53 mutation had a significantly shorter OS (all p < 0.05) in both circulating free DNA and tumor tissue. The mutations (no SNP) of NOTCH1 (all p < 0.05) significantly contributed to worse OS in the two cohorts.
Collapse
Affiliation(s)
- Yue Hou
- Department of Hematology, School of Medicine, Zhongda Hospital, Institute of Hematology Southeast University, Southeast University, Nanjing, China
| | - Jie Zi
- Department of Hematology, School of Medicine, Zhongda Hospital, Institute of Hematology Southeast University, Southeast University, Nanjing, China
| | - Shuo Liu
- Department of Hematology, School of Medicine, Zhongda Hospital, Institute of Hematology Southeast University, Southeast University, Nanjing, China
| | - Qinyu Ge
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Zheng Ge
- Department of Hematology, School of Medicine, Zhongda Hospital, Institute of Hematology Southeast University, Southeast University, Nanjing, China
| |
Collapse
|
5
|
Cost and clinical benefit of imaging surveillance after treatment for breast implant-associated anaplastic large cell lymphoma (BIA-ALCL). EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2021; 48:748-751. [PMID: 34974948 DOI: 10.1016/j.ejso.2021.12.463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/09/2021] [Accepted: 12/23/2021] [Indexed: 11/22/2022]
Abstract
Recent UK guidelines recommend that surveillance imaging should not be offered to patients who have undergone treatment for breast implant-associated anaplastic large cell lymphoma (BIA-ALCL) unless clinically indicated. The aim of this study was to explore the evolving practice at a tertiary referral unit and quantify the direct economic costs (DEC) associated with post-treatment BIA-ALCL routine radiological surveillance prior to adoption of the guidelines. Eleven patients were treated for BIA-ALCL between 2015 and 2020. At a median follow-up of 38 months (IQR 12-47) there were no local or distant relapses. Two patients did not have any radiological surveillance and 1 had follow-up elsewhere. The remaining 8 patients had a combination of positron emission tomography/computed tomography (PET/CT) (n = 10), CT (n = 2), breast ultrasound (n = 6), mammogram (n = 4) and breast magnetic resonance imaging (MRI) (n = 1) as routine imaging follow-up not guided by clinical concerns. Total cost of imaging was £10,396 (€12,257) with a median cost of £1953 (€2304) per patient [IQR £526-2029 (€621-2394)]. This cost could have been saved based on current guidelines recommending no routine surveillance for asymptomatic patients.
Collapse
|
6
|
Brice P, de Kerviler E, Friedberg JW. Classical Hodgkin lymphoma. Lancet 2021; 398:1518-1527. [PMID: 33493434 DOI: 10.1016/s0140-6736(20)32207-8] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 06/11/2020] [Accepted: 10/15/2020] [Indexed: 11/29/2022]
Abstract
Classical Hodgkin lymphoma is one of the more frequent lymphomas and is generally considered a highly curable disease with standard first-line chemotherapy and radiotherapy in some cases. Despite these outstanding results, major problems remain unresolved. First, there are still patients who will not be cured with front-line regimens and, second, many patients who are cured of classical Hodgkin lymphoma continue to die prematurely due to the late toxic effects of their therapy. Because the median age of patients with classical Hodgkin lymphoma is in the mid-30s, the disease's impact on the number of years lost from productive life is remarkable. In recent years, the gold standard of chemotherapy (often combined with radiotherapy) has changed, with the approval of immunotherapy mostly in relapse settings.
Collapse
Affiliation(s)
- Pauline Brice
- Department of Oncohaematology, Hôpital saint Louis APHP, Université Paris 7, Paris, France.
| | - Eric de Kerviler
- Department of Radiology, Hôpital saint Louis APHP, Université Paris 7, Paris, France
| | | |
Collapse
|
7
|
Ghesquières H, Rossi C, Cherblanc F, Le Guyader-Peyrou S, Bijou F, Sujobert P, Fabbro-Peray P, Bernier A, Belot A, Chartier L, Fornecker LM, Baldi I, Bouabdallah K, Laurent C, Oberic L, Morineau N, Le Gouill S, Morschhauser F, Haioun C, Damaj G, Guidez S, Labouré G, Fitoussi O, Lebras L, Gressin R, Salles G, Ysebaert L, Monnereau A. A French multicentric prospective prognostic cohort with epidemiological, clinical, biological and treatment information to improve knowledge on lymphoma patients: study protocol of the "REal world dAta in LYmphoma and survival in adults" (REALYSA) cohort. BMC Public Health 2021; 21:432. [PMID: 33653294 PMCID: PMC7927409 DOI: 10.1186/s12889-021-10433-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 02/12/2021] [Indexed: 12/24/2022] Open
Abstract
Background Age-adjusted lymphoma incidence rates continue to rise in France since the early 80’s, although rates have slowed since 2010 and vary across subtypes. Recent improvements in patient survival in major lymphoma subtypes at population level raise new questions about patient outcomes (i.e. quality of life, long-term sequelae). Epidemiological studies have investigated factors related to lymphoma risk, but few have addressed the extent to which socioeconomic status, social institutional context (i.e. healthcare system), social relationships, environmental context (exposures), individual behaviours (lifestyle) or genetic determinants influence lymphoma outcomes, especially in the general population. Moreover, the knowledge of the disease behaviour mainly obtained from clinical trials data is partly biased because of patient selection. Methods The REALYSA (“REal world dAta in LYmphoma and Survival in Adults”) study is a real-life multicentric cohort set up in French areas covered by population-based cancer registries to study the prognostic value of epidemiological, clinical and biological factors with a prospective 9-year follow-up. We aim to include 6000 patients over 4 to 5 years. Adult patients without lymphoma history and newly diagnosed with one of the following 7 lymphoma subtypes (diffuse large B-cell, follicular, marginal zone, mantle cell, Burkitt, Hodgkin, mature T-cell) are invited to participate during a medical consultation with their hematologist. Exclusion criteria are: having already received anti-lymphoma treatment (except pre-phase) and having a documented HIV infection. Patients are treated according to the standard practice in their center. Clinical data, including treatment received, are extracted from patients’ medical records. Patients’ risk factors exposures and other epidemiological data are obtained at baseline by filling out a questionnaire during an interview led by a clinical research assistant. Biological samples are collected at baseline and during treatment. A virtual tumor biobank is constituted for baseline tumor samples. Follow-up data, both clinical and epidemiological, are collected every 6 months in the first 3 years and every year thereafter. Discussion This cohort constitutes an innovative platform for clinical, biological, epidemiological and socio-economic research projects and provides an opportunity to improve knowledge on factors associated to outcome of lymphoma patients in real life. Trial registration 2018-A01332–53, ClinicalTrials.gov identifier: NCT03869619. Supplementary Information The online version contains supplementary material available at 10.1186/s12889-021-10433-4.
Collapse
Affiliation(s)
- Hervé Ghesquières
- Hospices Civils de Lyon, Lyon Sud Hospital, 165 Chemin du Grand Revoyet, 69310, Pierre-Bénite, France.
| | - Cédric Rossi
- CHU Dijon, 10 Boulevard Maréchal De Lattre De Tassigny, 21000, Dijon, France
| | - Fanny Cherblanc
- LYSARC, 165 Chemin du Grand Revoyet, 69310, Pierre-Bénite, France
| | | | - Fontanet Bijou
- Bergonié Institute, 229 Cours de l'Argonne, 33076, Bordeaux, France
| | - Pierre Sujobert
- Hospices Civils de Lyon, Lyon Sud Hospital, 165 Chemin du Grand Revoyet, 69310, Pierre-Bénite, France
| | | | - Adeline Bernier
- LYSARC, 165 Chemin du Grand Revoyet, 69310, Pierre-Bénite, France
| | - Aurélien Belot
- LYSARC, 165 Chemin du Grand Revoyet, 69310, Pierre-Bénite, France
| | - Loic Chartier
- LYSARC, 165 Chemin du Grand Revoyet, 69310, Pierre-Bénite, France
| | - Luc-Matthieu Fornecker
- Cancerology Institute Strasbourg Europe, Avenue Molière, BP 428, 67098, Strasbourg, France
| | - Isabelle Baldi
- Inserm U1219 - EPICENE team, Université de Bordeaux, Bordeaux, France
| | | | - Camille Laurent
- Toulouse Research Center in Cancerology, 2 Avenue Hubert Curien, 31037, Toulouse, France
| | - Lucie Oberic
- IUCT Oncopole, 1 Avenue Irène Joliot Curie, 31100, Toulouse, France
| | - Nadine Morineau
- CHD Vendée, Boulevard Stéphane Moreau, 85000, La Roche-sur-Yon, France
| | | | | | - Corinne Haioun
- Henri Mondor Hospital, 51 Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil, France
| | - Gandhi Damaj
- Hematology Institute of Basse Normandie, 6 Avenue Côte de Nacre, 14033, Caen, France
| | | | - Gaëlle Labouré
- CH Libourne, 112 Rue de la Marne, 33500, Libourne, France
| | - Olivier Fitoussi
- Polyclinique Bordeaux Nord Aquitaine, 15-35 Rue Claude Boucher, 33300, Bordeaux, France
| | - Laure Lebras
- Léon Bérard Center, 28 rue Laennec, 69008, Lyon, France
| | - Rémy Gressin
- CHU Grenoble, Bd de la Chantourne BP 217, 38043, Grenoble, France
| | - Gilles Salles
- Hospices Civils de Lyon, Lyon Sud Hospital, 165 Chemin du Grand Revoyet, 69310, Pierre-Bénite, France
| | - Loïc Ysebaert
- IUCT Oncopole, 1 Avenue Irène Joliot Curie, 31100, Toulouse, France
| | - Alain Monnereau
- Inserm U1219 - EPICENE team, Université de Bordeaux, Bordeaux, France
| |
Collapse
|
8
|
Osunkwo I, O'Connor HF, Saah E. Optimizing the management of chronic pain in sickle cell disease. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2020; 2020:562-569. [PMID: 33275672 PMCID: PMC7727591 DOI: 10.1182/hematology.2020000143] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Chronic pain in sickle cell disease (SCD) refers to pain present on most days lasting over six months. It can start during childhood and the prevalence increases with age. By adulthood, over 55% of patients experience pain on over 50% of days; 29% reporting pain on 95% of days. The true prevalence of chronic pain in SCD is likely underappreciated as it is mostly managed at home. Patients with chronic pain and SCD frequently seek acute care for exacerbation of underlying chronic pain difficult to distinguish from their usual acute vaso-occlusive crises. When treating chronic pain in SCD, the challenge is distinguishing between non-SCD related etiologies versus chronic pain resulting from SCD pathophysiological processes. This distinction is important to delineate as it will drive appropriate management strategies. Chronic pain in SCD has profound consequences for the patient; is often associated with comorbid psychiatric illnesses (depression and anxiety), not dissimilar from other chronic pain syndromes. They may also experience challenges with sleep hygiene, various somatic symptoms, and chronic fatigue that impair quality of life. How best to treat chronic pain in SCD is not definitively established. Both acute and chronic pain in SCD is typically treated with opioids. Emerging data suggests that chronic opioid therapy (COT) is a suboptimal treatment strategy for chronic pain. This review will discuss the complexity of managing chronic pain in SCD; pain that may be dependent or independent of the underlying SCD diagnosis. We will also describe alternative treatment approaches to high-dose COT.
Collapse
Affiliation(s)
| | | | - Elna Saah
- Children’s HealthCare of Atlanta, Emory University College of Medicine, Atlanta, GA
| |
Collapse
|
9
|
Ofori K, Bhagat G, Rai AJ. Exosomes and extracellular vesicles as liquid biopsy biomarkers in diffuse large B-cell lymphoma: Current state of the art and unmet clinical needs. Br J Clin Pharmacol 2020; 87:284-294. [PMID: 33080045 DOI: 10.1111/bcp.14611] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/15/2020] [Accepted: 09/21/2020] [Indexed: 12/15/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common type of non-Hodgkin's lymphoma, and it constitutes biologically heterogeneous entities. Standard first-line therapies cure ~60% of patients, the rest being either refractory or experiencing relapse. Currently, there are no robust predictive biomarkers of therapeutic response. Heterogeneity of DLBCL is partly explained by the cell of origin (COO), ie, germinal centre B cell or activated B cell, with the latter exhibiting worse prognosis. While gene expression profiling (GEP) is the gold standard for determining COO, surrogate immunohistochemical algorithms are used clinically, but show significant discordance with GEP. Recently, additional genetic subgroups with different prognoses have been reported. However, the tools/expertise required for analysis prohibit widespread deployment. Liquid biopsy-based assays show promise in providing clinically actionable information, are noninvasive and facilitate serial sampling to assess mechanisms of therapy resistance. Circulating, cell-free DNA analysis has shown enhanced sensitivity for detecting molecular alterations, but this modality cannot determine alterations of the tumor proteome or on signalling pathways. Exosomes are endosomally derived vesicles, are found in high abundance in body fluids and are readily isolated using a variety of methods. Tumour-derived exosomes can yield data regarding genetic, transcriptional, and proteomic changes useful for diagnosis, prognosis, and therapy of DLBCL. At present, standardized techniques for isolating exosomes are lacking and discriminating between exosomes from neoplastic and normal B cells is challenging. Refinements in isolation procedures are required to realize their full potential as precision medicine tools to provide comprehensive information on disease subtypes, identify prognostic factors, allow real-time monitoring of therapy response and delineate novel drug targets.
Collapse
Affiliation(s)
- Kenneth Ofori
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 622 West 168th Street, New York, NY, 10032
| | - Govind Bhagat
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 622 West 168th Street, New York, NY, 10032
| | - Alex J Rai
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 622 West 168th Street, New York, NY, 10032
| |
Collapse
|
10
|
Paltiel O, Raviv Sharabi G, Tzemach R, Rechavi T, Trachtenberg E, Goldschmidt N, Dann EJ, Bar-Shalom R. Limiting surveillance imaging for patients with lymphoma in remission: a mixed methods study leading to a Choosing Wisely recommendation. BMJ Qual Saf 2020; 30:300-310. [PMID: 32467340 DOI: 10.1136/bmjqs-2019-010756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/17/2020] [Accepted: 05/05/2020] [Indexed: 11/04/2022]
Abstract
BACKGROUND Under the 'Choosing Wisely' (CW) framework, professional organisations internationally have advocated limiting imaging for asymptomatic patients following curative cancer therapy, based on limited value and high cost. F18-fluorodeoxyglucose (FDG) positron emission tomography-CT (PET/CT) was widely adopted locally for surveillance lymphoma imaging after 2004. OBJECTIVES Prior to ratification of a local CW recommendation to limit surveillance imaging in lymphoma, we aimed to assess: (A) performance characteristics of surveillance FDG-PET/CT; (B) rates, clinical consequences and costs of false positives (FP); and (C) patients and professionals' attitudes towards overuse. METHODS Mixed methods (quantitative and qualitative) study. We analysed surveillance FDG-PET/CT results of two patient cohorts (n1=215 Hodgkin lymphoma and non-Hodgkin lymphoma; n2=203 Hodgkin lymphoma only). FPs were defined by negative biopsy or clinical follow-up. We held focus group discussions and in-depth interviews eliciting attitudes of 26 patients and 11 clinicians, respectively. RESULTS FPs were observed in 25.1% (95% CI 20.5 to 30.5) per scan-cohort 1, and 41.7% (95% CI 37.9 to 45.6) per patient-cohort 2, engendering frequent additional testing. Specific characteristics and location of findings altered the FP rate. The estimated cost per relapse detected was $50 000 (cohort 2). Patients sought reassurance via surveillance imaging, which they considered highly accurate, yet stressful. Aware of radiation risks, they were largely unconcerned about consequences of FPs. Confidence in the treating physicians was an important factor in patients' acceptance of forgoing imaging. Clinicians, frequently under patient pressure to order imaging, generally believed that it did not affect prognosis (with important exceptions), welcomed professional guidelines, but rejected regulatory restrictions on its use. CONCLUSION Acceptance of CW recommendations to limit overuse may be enhanced by quantitative data on consequences and costs of surveillance imaging, supplemented by qualitative data on patient and physician attitudes.
Collapse
Affiliation(s)
- Ora Paltiel
- Braun School of Public Health and Community Medicine, Hadassah-Hebrew University of Jerusalem, Jerusalem, Israel .,Hematology, Hadassah Medical Center, Jerusalem, Israel
| | | | - Reut Tzemach
- Rheumatology, Tel Aviv Ichilov-Sourasky Medical Center, Tel Aviv, Israel
| | - Talya Rechavi
- Braun School of Public Health and Community Medicine, Hadassah-Hebrew University of Jerusalem, Jerusalem, Israel
| | - Estherina Trachtenberg
- Hematology, Rambam Hospital, Haifa, Israel.,Rappaport School of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | | | - Eldad J Dann
- Hematology, Rambam Hospital, Haifa, Israel.,Rappaport School of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | | |
Collapse
|
11
|
Glober G, Gunther J, Fang P, Milgrom S, Korivi BR, Jensen CT, Wagner-Bartak NA, Ahmed S, Lee HJ, Nair R, Steiner R, Parmar S, Iyer S, Westin J, Fayad L, Rodriguez MA, Neelapu S, Nastoupil L, Flowers CR, Dabaja BS, Pinnix CC. Imaging Surveillance of Limited-stage Classic Hodgkin Lymphoma Patients After PET-CT-documented First Remission. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:533-541. [PMID: 32291233 PMCID: PMC10071957 DOI: 10.1016/j.clml.2020.02.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/03/2020] [Accepted: 02/09/2020] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Early stage Hodgkin lymphoma (ESHL) is highly curable; however, 10% to 15% of patients experience relapse. We examined the utilization of follow-up imaging for patients with ESHL who achieved a metabolic complete response after upfront therapy. MATERIALS AND METHODS The records of adult patients treated at a single institution between 2003 and 2014 were reviewed. Positron emission tomography-computed tomography (PET-CT) and CT scan frequency was quantified during the 2 years following treatment and subsequent visits beyond 2 years. RESULTS The study cohort contained 179 patients. The median age was 31 years; bulky disease was present in 30%. ABVD (doxorubicin, bleomycin, vinblastine, and dacarbazine) or AVD (doxorubicin, vinblastine, and dacarbazine) was given in 97%; 75% received radiation therapy. At a median follow-up of 6.9 years, the 5-year progression-free and overall survival rates were 93.7% and 98.1%, respectively. Relapse occurred in 5% (n = 9) of patients at a median of 9.1 months (range, 4.6-27.2 months) from therapy. Two patients presented with symptoms prompting imaging in follow-up. Within 2 years after therapy, 376 PET-CT scans and 3325 CT scans were performed, yielding an average of 2.1 PET-CTs and 18.6 CTs per patient. Of the initial 179 patients, 113 had follow-up conducted beyond 2 years post-therapy; an average of 2.7 PET-CTs and 33.2 CTs were performed. In the 2-year post-therapy period, 463 scans were performed per relapse detected. CONCLUSION In this cohort of patients with ESHL who responded completely to frontline therapy, the relapse rate was low. Routine imaging surveillance lacks clinical benefit in this patient population.
Collapse
Affiliation(s)
- Gordon Glober
- University of Central Florida College of Medicine, Orlando, FL
| | - Jillian Gunther
- Departments of Radiation Oncology, MD Anderson Cancer Center, Houston, TX
| | - Penny Fang
- Departments of Radiation Oncology, MD Anderson Cancer Center, Houston, TX
| | - Sarah Milgrom
- Department of Radiation Oncology, University of Colorado, Denver, CO
| | - Brinda Rao Korivi
- Department of Diagnostic Radiology, MD Anderson Cancer Center, Houston, TX
| | - Corey T Jensen
- Department of Diagnostic Radiology, MD Anderson Cancer Center, Houston, TX
| | | | - Sairah Ahmed
- Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX
| | - Hun Ju Lee
- Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX
| | - Ranjit Nair
- Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX
| | - Raphael Steiner
- Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX
| | - Simrit Parmar
- Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX
| | - Swaminathan Iyer
- Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX
| | - Jason Westin
- Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX
| | - Luis Fayad
- Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX
| | - M Alma Rodriguez
- Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX
| | - Sattva Neelapu
- Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX
| | - Loretta Nastoupil
- Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX
| | | | - Bouthaina S Dabaja
- Departments of Radiation Oncology, MD Anderson Cancer Center, Houston, TX
| | - Chelsea C Pinnix
- Departments of Radiation Oncology, MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
12
|
Kallam A, Adusumalli J, Armitage JO. Surveillance in Patients With Diffuse Large B Cell Lymphoma. Mayo Clin Proc 2020; 95:157-163. [PMID: 31902411 DOI: 10.1016/j.mayocp.2019.05.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/03/2019] [Accepted: 05/14/2019] [Indexed: 01/28/2023]
Abstract
With improvement in the cure rates for diffuse large B cell lymphoma, the question of surveillance imaging in patients who achieve complete remission after the initial therapy has become relevant. Some of the clinical practice guidelines recommend surveillance scanning. However, several studies have reported no benefit in overall survival with scans. Moreover, studies have highlighted an increased risk for developing secondary malignancies because of exposure to ionizing radiation from the scans. Different international societies have contrasting guidelines for the role of surveillance computerized tomography scans in patients who achieve complete remission after first-line therapy. Any benefit of surveillance imaging must be balanced by the costs, risk of radiation exposure, and lack of survival benefit. The PubMed platform was searched using relevant keywords for English-language articles with no date restrictions. Search terms were cross-referenced with review articles, and additional articles were identified by manually searching reference lists. Results were reviewed by the authors and selected for inclusion based on relevance. We present a review of this current data available for surveillance imaging in patients with diffuse large B cell lymphoma.
Collapse
Affiliation(s)
- Avyakta Kallam
- Division of Oncology/Hematology, University of Nebraska Medical Center, Omaha.
| | | | - James O Armitage
- Division of Oncology/Hematology, University of Nebraska Medical Center, Omaha
| |
Collapse
|
13
|
Maurer MJ, Habermann TM, Shi Q, Schmitz N, Cunningham D, Pfreundschuh M, Seymour JF, Jaeger U, Haioun C, Tilly H, Ghesquieres H, Merli F, Ziepert M, Herbrecht R, Flament J, Fu T, Flowers CR, Coiffier B. Progression-free survival at 24 months (PFS24) and subsequent outcome for patients with diffuse large B-cell lymphoma (DLBCL) enrolled on randomized clinical trials. Ann Oncol 2019; 29:1822-1827. [PMID: 29897404 DOI: 10.1093/annonc/mdy203] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Patients with diffuse large B-cell lymphoma treated with first-line anthracycline-based immunochemotherapy and remaining in remission at 2 years have excellent outcomes. This study assessed overall survival (OS) stratified by progression-free survival (PFS) at 24 months (PFS24) using individual patient data from patients with DLBCL enrolled in multi-center, international randomized clinical trials as part of the Surrogate Endpoint for Aggressive Lymphoma (SEAL) Collaboration. Patients and methods PFS24 was defined as being alive and PFS24 after study entry. OS from PFS24 was defined as time from identified PFS24 status until death due to any cause. OS was compared with each patient's age-, sex-, and country-matched general population using expected survival and standardized mortality ratios (SMRs). Results A total of 5853 patients enrolled in trials in the SEAL database received rituximab as part of induction therapy and were included in this analysis. The median age was 62 years (range 18-92), and 56% were greater than 60 years of age. At a median follow-up of 4.4 years, 1337 patients (23%) had disease progression, 1489 (25%) had died, and 5101 had sufficient follow-up to evaluate PFS24. A total of 1423 assessable patients failed to achieve PFS24 with a median OS of 7.2 months (95% CI 6.8-8.1) after progression; 5-year OS after progression was 19% and SMR was 32.1 (95% CI 30.0-34.4). A total of 3678 patients achieved PFS24; SMR after achieving PFS24 was 1.22 (95% CI 1.09-1.37). The observed OS versus expected OS at 3, 5, and 7 years after achieving PFS24 was 93.1% versus 94.4%, 87.6% versus 89.5%, and 80.0% versus 83.7%, respectively. Conclusion Patients treated with rituximab containing anthracycline-based immunochemotherapy on clinical trials who are alive without progression at 24 months from the onset of initial therapy have excellent outcomes with survival that is marginally lower but clinically indistinguishable from the age-, sex-, and country-matched background population for 7 years after achieving PFS24.
Collapse
Affiliation(s)
- M J Maurer
- Department of Health Sciences Research, Mayo Clinic, Rochester, USA.
| | | | - Q Shi
- Department of Health Sciences Research, Mayo Clinic, Rochester, USA
| | - N Schmitz
- Department of Hematology, Oncology and Stem Cell Transplantation, Asklepios Hospital St. Georg, Hamburg, Germany
| | - D Cunningham
- Department of Medicine, The Royal Marsden Hospital, Surrey, UK
| | - M Pfreundschuh
- Internal Medicine I, University of the Saarland, Homberg, Germany
| | - J F Seymour
- Peter MacCallum Cancer Centre, Melbourne, Australia
| | - U Jaeger
- Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - C Haioun
- Lymphoid Malignancies Unit, AP-HP Hôpital Henri Mondor, Créteil, France
| | - H Tilly
- Henri Becquerel Centre, University of Rouen, Rouen, France
| | - H Ghesquieres
- Department of Hematology, Centre Hospitalier Lyon-Sud, Pierre-Benite, France
| | - F Merli
- Hematology, Azienda Ospedaliera Santa Maria Nuova-IRCCS, Reggio Emilia, Italy
| | - M Ziepert
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - R Herbrecht
- Department of Oncology and Hematology, Hôpital de Hautepierre, Strasbourg, France
| | - J Flament
- Celgene Corporation, Boudry, Switzerland
| | - T Fu
- Celgene Corporation, Summit
| | - C R Flowers
- Department of Bone Marrow and Stem Cell Transplantation, Winship Cancer Institute of Emory University, Atlanta, USA
| | - B Coiffier
- Department of Hematology, Centre Hospitalier Lyon-Sud, Pierre-Benite, France
| |
Collapse
|
14
|
Harkins RA, Patel SP, Flowers CR. Cost burden of diffuse large B-cell lymphoma. Expert Rev Pharmacoecon Outcomes Res 2019; 19:645-661. [PMID: 31623476 PMCID: PMC6930962 DOI: 10.1080/14737167.2019.1680288] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 10/11/2019] [Indexed: 12/15/2022]
Abstract
Introduction: Diffuse large B-cell lymphoma (DLBCL) is the most common non-Hodgkin lymphoma and is a clinically heterogeneous disease. Treatment pathways for DLBCL are diverse and integrate established and novel therapies.Areas covered: We review the cost burden of DLBCL and the cost-effectiveness of DLBCL management including precision and cellular medicine. We utilized Medical Subject Heading (MeSH) terms and keywords to search the National Library of Medicine online MEDLINE database (PubMed) for articles related to cost, cost burden, and cost-of-illness of DLBCL and cost-effectiveness of DLBCL management strategies published in English as of June 2019.Expert commentary: Available and developing DLBCL therapies offer improved outcomes and often curative treatment at considerable financial expense, and the total cost burden for DLBCL management is substantial for patients and the healthcare system. In the era of personalized medicine, CAR T cells and targeted therapies provide exciting avenues for current and future DLBCL care and can further increase treatment cost. Determinations of cost and cost-effectiveness in DLBCL treatment pathways should continue to guide care providers and systems in identifying cost reduction strategies to provide appropriate therapies to the greatest number of patients in treating DLBCL.
Collapse
Affiliation(s)
- R Andrew Harkins
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Sharvil P Patel
- Department of Quantitative Theories and Methods, Emory University, Atlanta, GA, USA
| | - Christopher R Flowers
- Department of Hematology and Oncology, Winship Research Informatics Shared Resource Emory University School of Medicine Winship Cancer Institute, Atlanta, GA, USA
| |
Collapse
|
15
|
Harkins RA, Chang A, Patel SP, Lee MJ, Goldstein JS, Merdan S, Flowers CR, Koff JL. Remaining challenges in predicting patient outcomes for diffuse large B-cell lymphoma. Expert Rev Hematol 2019; 12:959-973. [PMID: 31513757 PMCID: PMC6821591 DOI: 10.1080/17474086.2019.1660159] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 08/22/2019] [Indexed: 12/28/2022]
Abstract
Introduction: Diffuse large B-cell lymphoma (DLBCL) is the most common non-Hodgkin lymphoma and is an aggressive malignancy with heterogeneous outcomes. Diverse methods for DLBCL outcomes assessment ranging from clinical to genomic have been developed with variable predictive and prognostic success.Areas covered: The authors provide an overview of the various methods currently used to estimate prognosis in DLBCL patients. Models incorporating cell of origin, genomic features, sociodemographic factors, treatment effectiveness measures, and machine learning are described.Expert opinion: The clinical and genetic heterogeneity of DLBCL presents distinct challenges in predicting response to therapy and overall prognosis. Successful integration of predictive and prognostic tools in clinical trials and in a standard clinical workflow for DLBCL will likely require a combination of methods incorporating clinical, sociodemographic, and molecular factors with the aid of machine learning and high-dimensional data analysis.
Collapse
Affiliation(s)
- R. Andrew Harkins
- Emory University School of Medicine, Atlanta, Georgia 30322-1007, USA
| | - Andres Chang
- Winship Cancer Institute of Emory University, Atlanta, Georgia 30322-1007, USA
| | | | - Michelle J. Lee
- Emory University School of Medicine, Atlanta, Georgia 30322-1007, USA
| | | | - Selin Merdan
- Winship Cancer Institute of Emory University, Atlanta, Georgia 30322-1007, USA
- Georgia Institute of Technology, Atlanta, Georgia 30332-0002, USA
| | | | - Jean L. Koff
- Winship Cancer Institute of Emory University, Atlanta, Georgia 30322-1007, USA
| |
Collapse
|
16
|
Stopsack KH, Cerhan JR. Cumulative Doses of Ionizing Radiation From Computed Tomography: A Population-Based Study. Mayo Clin Proc 2019; 94:2011-2021. [PMID: 31248696 PMCID: PMC6778511 DOI: 10.1016/j.mayocp.2019.05.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 01/21/2023]
Abstract
OBJECTIVE To assess cumulative radiation doses from computed tomography (CT), patient characteristics, and clinical indications for CT in a population-based sample. PATIENTS AND METHODS A cohort study using medical records linkage through the Rochester Epidemiology Project was conducted to ascertain all CT examinations in Olmsted County, Minnesota, performed between January 1, 2004, and December 31, 2013, among all adults who were alive for 3 or more years after the end of follow-up (to exclude exposures preceding death). Ten-year cumulative effective ionizing radiation doses were estimated on the basis of typical doses per CT modality. Among patients with high doses (≥100 mSv/10 years), CT scans were reviewed for clinical setting, indications, and results. RESULTS Of 54,447 adults (median age, 44.0 years at inclusion), 26,377 (48.4%) underwent at least one CT. Ten-year radiation doses from CT were 0.1 to 9.9 mSv in 15.8% of the population (8593 patients), 10 to 24.9 mSv in 16.9% (9502), 25 to 99.9 mSv in 13.8% (7492), and 100 mSv or greater in 1.9% (1041). Computed tomography of the abdomen and pelvis accounted for 67.2% of the estimated dose. In multivariable models, doses differed 1.21-fold to 2.16-fold between extreme categories of age, body mass index, education level, smoking status, and by race. Of 600 CTs in 200 patients with high doses, 70.5% were obtained for restaging of solid cancers and lymphoma, abdominal pain, infection, kidney stones, follow-up of nodules or masses, and chest pain/evaluation for pulmonary embolism. CONCLUSION Exposure to ionizing radiation from CT occurred disproportionally in specific subgroups of the population. A limited number of clinical indications contributed the majority of radiation among adults with high doses.
Collapse
Affiliation(s)
- Konrad H Stopsack
- Department of Internal Medicine, Mayo Clinic, Rochester, MN; Department of Health Sciences Research, Mayo Clinic, Rochester, MN.
| | - James R Cerhan
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| |
Collapse
|
17
|
Singh A, Nandwana V, Rink JS, Ryoo SR, Chen TH, Allen SD, Scott EA, Gordon LI, Thaxton CS, Dravid VP. Biomimetic Magnetic Nanostructures: A Theranostic Platform Targeting Lipid Metabolism and Immune Response in Lymphoma. ACS NANO 2019; 13:10301-10311. [PMID: 31487458 DOI: 10.1021/acsnano.9b03727] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
B-cell lymphoma cells depend upon cholesterol to maintain pro-proliferation and pro-survival signaling via the B-cell receptor. Targeted cholesterol depletion of lymphoma cells is an attractive therapeutic strategy. We report here high-density lipoprotein mimicking magnetic nanostructures (HDL-MNSs) that can bind to the high-affinity HDL receptor, scavenger receptor type B1 (SR-B1), and interfere with cholesterol flux mechanisms in SR-B1 receptor positive lymphoma cells, causing cellular cholesterol depletion. In addition, the MNS core can be utilized for its ability to generate heat under an external radio frequency field. The thermal activation of MNS can lead to both innate and adaptive antitumor immune responses by inducing the expression of heat shock proteins that lead to activation of antigen presenting cells and finally lymphocyte trafficking. In the present study, we demonstrate SR-B1 receptor mediated binding and cellular uptake of HDL-MNS and prevention of phagolysosome formation by transmission electron microscopy, fluorescence microscopy, and ICP-MS analysis. The combinational therapeutics of cholesterol depletion and thermal activation significantly improves therapeutic efficacy in SR-B1 expressing lymphoma cells. HDL-MNS reduces the T2 relaxation time under magnetic resonance imaging (MRI) more effectively compared with a commercially available contrast agent, and the specificity of HDL-MNS toward the SR-B1 receptor leads to differential contrast between SR-B1 positive and negative cells suggesting its utility in diagnostic imaging. Overall, we have demonstrated that HDL-MNSs have cell specific targeting efficiency, can modulate cholesterol efflux, can induce thermal activation mediated antitumor immune response, and possess high contrast under MRI, making it a promising theranostic platform in lymphoma.
Collapse
Affiliation(s)
- Abhalaxmi Singh
- Department of Materials Science & Engineering , Northwestern University , Evanston , Illinois 60208 , United States
- International Institute of Nanotechnology , Evanston , Illinois 60208 , United States
| | - Vikas Nandwana
- Department of Materials Science & Engineering , Northwestern University , Evanston , Illinois 60208 , United States
- International Institute of Nanotechnology , Evanston , Illinois 60208 , United States
| | - Jonathan S Rink
- Department of Medicine, Division of Hematology/Oncology, Feinberg School of Medicine , Northwestern University , Chicago , Illinois 60611 , United States
- Simpson-Querrey Institute for Bionanotechnology , Northwestern University , Chicago , Illinois 60611 , United States
| | - Soo-Ryoon Ryoo
- Department of Materials Science & Engineering , Northwestern University , Evanston , Illinois 60208 , United States
- International Institute of Nanotechnology , Evanston , Illinois 60208 , United States
| | - Tzu Hung Chen
- Department of Materials Science & Engineering , Northwestern University , Evanston , Illinois 60208 , United States
| | - Sean David Allen
- Interdisciplinary Biological Sciences Program , Northwestern University , Evanston , Illinois 60208 , United States
| | - Evan A Scott
- Simpson-Querrey Institute for Bionanotechnology , Northwestern University , Chicago , Illinois 60611 , United States
- Interdisciplinary Biological Sciences Program , Northwestern University , Evanston , Illinois 60208 , United States
- Department of Biomedical Engineering , Northwestern University , Evanston , Illinois 60208 , United States
| | - Leo I Gordon
- Department of Medicine, Division of Hematology/Oncology, Feinberg School of Medicine , Northwestern University , Chicago , Illinois 60611 , United States
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University , Chicago , Illinois 60611 , United States
| | - C Shad Thaxton
- International Institute of Nanotechnology , Evanston , Illinois 60208 , United States
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University , Chicago , Illinois 60611 , United States
- Department of Urology, Feinberg School of Medicine , Northwestern University , Chicago , Illinois 60611 , United States
| | - Vinayak P Dravid
- Department of Materials Science & Engineering , Northwestern University , Evanston , Illinois 60208 , United States
- International Institute of Nanotechnology , Evanston , Illinois 60208 , United States
| |
Collapse
|
18
|
Çağlayan Ç, Goldstein JS, Ayer T, Rai A, Flowers CR. A population-based multistate model for diffuse large B-cell lymphoma-specific mortality in older patients. Cancer 2019; 125:1837-1847. [PMID: 30707765 PMCID: PMC6509004 DOI: 10.1002/cncr.31981] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/17/2018] [Accepted: 12/07/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Despite effective therapies, outcomes for diffuse large B-cell lymphoma (DLCBL) remain heterogeneous in older individuals due to comorbid diseases and variations in disease biology. METHODS Using the Surveillance, Epidemiology, and End Results (SEER)-Medicare database, the authors conducted a multistate survival analysis of 11,780 patients with DLBCL who were aged ≥65 years at the time of diagnosis (2002-2009). Cox proportional hazards models were used to specify the impact of prognostic factors on overall survival and cause-specific deaths, and the Aalen-Johansen estimator was used to project the course of DLBCL over time with or without standard therapy with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP). RESULTS Advanced age (hazard ratio [HR] for ages 71-75 years: 1.25; HR for ages 76-80 years: 1.46; HR for ages 81-85 years: 1.88; and HR for age ≥86 years: 2.26), DLBCL stage (HR for Ann Arbor stage II: 1.28; HR for stage III: 1.54; and HR for stage IV: 1.95), Charlson Comorbidity Index (CCI) ≥1 (HR for CCI of 1, 1.15; and HR for CCI >1, 1.37), and not being married (HR, 1.12) were associated with an increased risk of DLBCL-specific death. Being female (HR, 0.91) and of higher socioeconomic status (HR, 0.91) were associated with a lower risk of DLBCL-related mortality after therapy. For patients treated with R-CHOP (3610 patients), the risk of death due to DLBCL was 14.0% and 18.6%, respectively, at 2 and 5 years of treatment and plateaued afterward, confirming a 5-year "cure" point while receiving R-CHOP among older patients. CONCLUSIONS Conducting a survival analysis over a large data set, the current study evaluated competing risks for death within a multistate modeling framework, and identified age, sex, and CCI as risk factors for DLBCL-specific and other causes of death.
Collapse
Affiliation(s)
- Çağlar Çağlayan
- H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, Georgia
| | | | - Turgay Ayer
- H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, Georgia
| | - Ashish Rai
- Outcomes Research, Surveillance and Health Services Research Program, American Cancer Society Inc, Atlanta, Georgia
| | | |
Collapse
|
19
|
Crombie J, Armand P. The Emerging Role of Liquid Biopsies in Lymphoproliferative Disorders. Curr Hematol Malig Rep 2019; 14:11-21. [DOI: 10.1007/s11899-019-0493-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
20
|
Shi Q, Flowers CR. Reply to H.J.A. Adams et al. J Clin Oncol 2019; 37:526-527. [PMID: 30592641 DOI: 10.1200/jco.18.02022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Qian Shi
- Qian Shi, PhD, Mayo Clinic, Rochester, MN; and Christopher R. Flowers, MD, Emory University, Atlanta, GA
| | - Christopher R Flowers
- Qian Shi, PhD, Mayo Clinic, Rochester, MN; and Christopher R. Flowers, MD, Emory University, Atlanta, GA
| |
Collapse
|
21
|
Do Deauville Scores Improve the Clinical Utility of End-of-Therapy FDG PET Scans for Pediatric Hodgkin Lymphoma? AJR Am J Roentgenol 2018; 212:456-460. [PMID: 30476458 DOI: 10.2214/ajr.18.19755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE The purpose of this study was to evaluate the clinical utility of Deauville scores in interpretation of end-of-chemotherapy FDG PET scans. CONCLUSION Deauville scores improve the clinical utility of end-of-chemotherapy PET, as evidenced by an increase in positive predictive value to 72.7% from 44.4% on the basis of report alone. The negative predictive value remains greater than 95%.
Collapse
|
22
|
Wu FT, Lu L, Xu W, Li JY. Circulating tumor DNA: clinical roles in diffuse large B cell lymphoma. Ann Hematol 2018; 98:255-269. [PMID: 30368587 DOI: 10.1007/s00277-018-3529-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 10/17/2018] [Indexed: 12/16/2022]
Abstract
Diffuse large B cell lymphoma (DLBCL), the most common non-Hodgkin lymphoma (NHL), is a clinically and molecularly heterogeneous malignant lymphoproliferative disease. In the era of personalized medicine, genetic information is critical to early diagnosis, aiding risk stratification, directing therapeutic option, and monitoring disease relapse. However, lacking a circulating disease with most DLBCL cases hampers the acquisition of tumor genomic landscapes and disease dynamics. Circulating tumor DNA (ctDNA) is a novel noninvasive, real-time, and tumor-specific biomarker, reliably reflecting the comprehensive tumor genetic profiles, thus holds great promise in individualized medicine, including precise diagnosis and prognosis, response monitoring, and relapse detection of DLBCL. Here, we reviewed the recent advances of ctDNA in DLBCL and discussed its clinical values at different time points during the disease courses by comparing with the current routine methods in clinical practice. Collectively, we anticipated that ctDNA will ultimately be integrated into the management of DLBCL to facilitate precision medicine.
Collapse
Affiliation(s)
- Fang-Tian Wu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Luo Lu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Wei Xu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China.
| | - Jian-Yong Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China.
| |
Collapse
|
23
|
Mullen EA, Chi YY, Hibbitts E, Anderson JR, Steacy KJ, Geller JI, Green DM, Khanna G, Malogolowkin MH, Grundy PE, Fernandez CV, Dome JS. Impact of Surveillance Imaging Modality on Survival After Recurrence in Patients With Favorable-Histology Wilms Tumor: A Report From the Children's Oncology Group. J Clin Oncol 2018; 36:JCO1800076. [PMID: 30335557 PMCID: PMC6269130 DOI: 10.1200/jco.18.00076] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PURPOSE The use of computed tomography (CT) for routine surveillance to detect recurrence in patients with Wilms tumor (WT) has increased in recent years. The utility of CT, despite increased risk and cost, to improve outcome for these patients is unknown. We conducted a retrospective analysis with patients enrolled in the fifth National Wilms Tumor Study (NWTS-5) to determine if surveillance with CT correlates with improved overall survival (OS) after recurrence compared with chest x-ray (CXR) and abdominal ultrasound (US). PATIENTS AND METHODS Overall, 281 patients with recurrent unilateral favorable-histology WT were reviewed to assess how WT recurrence was detected: sign/symptoms (SS), surveillance imaging (SI) with CT scan, or SI with CXR/US. RESULTS The estimated 5-year OS rate after relapse was 67% (95% CI, 61% to 72%). Twenty-five percent of recurrences were detected with SS; 48.5%, with CXR/US; and 26.5%, with CT. Patients with SS had a 5-year OS rate of 59% (95% CI, 46% to 72%) compared with 70% (95% CI, 63% to 77%; P = .23) for those detected by SI. Recurrences detected by CT had a shorter median time from diagnosis to recurrence (0.60 years) compared with SS (0.91 years) or CXR/US (0.86 years; P = .003). For recurrences detected by SI, more tumor foci at relapse ( P < .001) and size of the largest focus greater than 2 cm ( P = .02) were associated with inferior OS. However, there was no difference in OS after relapse when recurrence was detected by CT versus CXR/US (5-year OS rate, 65% v 73%; P = .20). CONCLUSION In patients with favorable-histology WT, elimination of CT scans from surveillance programs is unlikely to compromise survival but would result in substantial reduction in radiation exposure and health care costs.
Collapse
Affiliation(s)
- Elizabeth A. Mullen
- Elizabeth A. Mullen, Dana-Farber Cancer Institute/Boston Children’s Cancer and Blood Disorders Center, Boston, MA; Yueh-Yun Chi and Emily Hibbitts, University of Florida, Gainesville, FL; James R. Anderson, Merck Research Laboratories, North Wales, PA; Katarina J. Steacy, University of Maryland Medical Center, Baltimore, MD; James I. Geller, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Daniel M. Green, St Jude Children’s Research Hospital, Memphis, TN; Geetika Khanna, Washington University School of Medicine, St Louis, MO; Marcio H. Malogolowkin, University of California at Davis Comprehensive Cancer Center, Sacramento, CA; Paul E. Grundy, Stollery Children’s Hospital, University of Alberta, Alberta; Conrad V. Fernandez, IWK Health Center, Dalhousie University Halifax, Nova Scotia, Canada; and Jeffrey S. Dome, Children’s National Health System, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Yueh-Yun Chi
- Elizabeth A. Mullen, Dana-Farber Cancer Institute/Boston Children’s Cancer and Blood Disorders Center, Boston, MA; Yueh-Yun Chi and Emily Hibbitts, University of Florida, Gainesville, FL; James R. Anderson, Merck Research Laboratories, North Wales, PA; Katarina J. Steacy, University of Maryland Medical Center, Baltimore, MD; James I. Geller, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Daniel M. Green, St Jude Children’s Research Hospital, Memphis, TN; Geetika Khanna, Washington University School of Medicine, St Louis, MO; Marcio H. Malogolowkin, University of California at Davis Comprehensive Cancer Center, Sacramento, CA; Paul E. Grundy, Stollery Children’s Hospital, University of Alberta, Alberta; Conrad V. Fernandez, IWK Health Center, Dalhousie University Halifax, Nova Scotia, Canada; and Jeffrey S. Dome, Children’s National Health System, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Emily Hibbitts
- Elizabeth A. Mullen, Dana-Farber Cancer Institute/Boston Children’s Cancer and Blood Disorders Center, Boston, MA; Yueh-Yun Chi and Emily Hibbitts, University of Florida, Gainesville, FL; James R. Anderson, Merck Research Laboratories, North Wales, PA; Katarina J. Steacy, University of Maryland Medical Center, Baltimore, MD; James I. Geller, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Daniel M. Green, St Jude Children’s Research Hospital, Memphis, TN; Geetika Khanna, Washington University School of Medicine, St Louis, MO; Marcio H. Malogolowkin, University of California at Davis Comprehensive Cancer Center, Sacramento, CA; Paul E. Grundy, Stollery Children’s Hospital, University of Alberta, Alberta; Conrad V. Fernandez, IWK Health Center, Dalhousie University Halifax, Nova Scotia, Canada; and Jeffrey S. Dome, Children’s National Health System, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - James R. Anderson
- Elizabeth A. Mullen, Dana-Farber Cancer Institute/Boston Children’s Cancer and Blood Disorders Center, Boston, MA; Yueh-Yun Chi and Emily Hibbitts, University of Florida, Gainesville, FL; James R. Anderson, Merck Research Laboratories, North Wales, PA; Katarina J. Steacy, University of Maryland Medical Center, Baltimore, MD; James I. Geller, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Daniel M. Green, St Jude Children’s Research Hospital, Memphis, TN; Geetika Khanna, Washington University School of Medicine, St Louis, MO; Marcio H. Malogolowkin, University of California at Davis Comprehensive Cancer Center, Sacramento, CA; Paul E. Grundy, Stollery Children’s Hospital, University of Alberta, Alberta; Conrad V. Fernandez, IWK Health Center, Dalhousie University Halifax, Nova Scotia, Canada; and Jeffrey S. Dome, Children’s National Health System, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Katarina J. Steacy
- Elizabeth A. Mullen, Dana-Farber Cancer Institute/Boston Children’s Cancer and Blood Disorders Center, Boston, MA; Yueh-Yun Chi and Emily Hibbitts, University of Florida, Gainesville, FL; James R. Anderson, Merck Research Laboratories, North Wales, PA; Katarina J. Steacy, University of Maryland Medical Center, Baltimore, MD; James I. Geller, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Daniel M. Green, St Jude Children’s Research Hospital, Memphis, TN; Geetika Khanna, Washington University School of Medicine, St Louis, MO; Marcio H. Malogolowkin, University of California at Davis Comprehensive Cancer Center, Sacramento, CA; Paul E. Grundy, Stollery Children’s Hospital, University of Alberta, Alberta; Conrad V. Fernandez, IWK Health Center, Dalhousie University Halifax, Nova Scotia, Canada; and Jeffrey S. Dome, Children’s National Health System, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - James I. Geller
- Elizabeth A. Mullen, Dana-Farber Cancer Institute/Boston Children’s Cancer and Blood Disorders Center, Boston, MA; Yueh-Yun Chi and Emily Hibbitts, University of Florida, Gainesville, FL; James R. Anderson, Merck Research Laboratories, North Wales, PA; Katarina J. Steacy, University of Maryland Medical Center, Baltimore, MD; James I. Geller, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Daniel M. Green, St Jude Children’s Research Hospital, Memphis, TN; Geetika Khanna, Washington University School of Medicine, St Louis, MO; Marcio H. Malogolowkin, University of California at Davis Comprehensive Cancer Center, Sacramento, CA; Paul E. Grundy, Stollery Children’s Hospital, University of Alberta, Alberta; Conrad V. Fernandez, IWK Health Center, Dalhousie University Halifax, Nova Scotia, Canada; and Jeffrey S. Dome, Children’s National Health System, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Daniel M. Green
- Elizabeth A. Mullen, Dana-Farber Cancer Institute/Boston Children’s Cancer and Blood Disorders Center, Boston, MA; Yueh-Yun Chi and Emily Hibbitts, University of Florida, Gainesville, FL; James R. Anderson, Merck Research Laboratories, North Wales, PA; Katarina J. Steacy, University of Maryland Medical Center, Baltimore, MD; James I. Geller, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Daniel M. Green, St Jude Children’s Research Hospital, Memphis, TN; Geetika Khanna, Washington University School of Medicine, St Louis, MO; Marcio H. Malogolowkin, University of California at Davis Comprehensive Cancer Center, Sacramento, CA; Paul E. Grundy, Stollery Children’s Hospital, University of Alberta, Alberta; Conrad V. Fernandez, IWK Health Center, Dalhousie University Halifax, Nova Scotia, Canada; and Jeffrey S. Dome, Children’s National Health System, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Geetika Khanna
- Elizabeth A. Mullen, Dana-Farber Cancer Institute/Boston Children’s Cancer and Blood Disorders Center, Boston, MA; Yueh-Yun Chi and Emily Hibbitts, University of Florida, Gainesville, FL; James R. Anderson, Merck Research Laboratories, North Wales, PA; Katarina J. Steacy, University of Maryland Medical Center, Baltimore, MD; James I. Geller, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Daniel M. Green, St Jude Children’s Research Hospital, Memphis, TN; Geetika Khanna, Washington University School of Medicine, St Louis, MO; Marcio H. Malogolowkin, University of California at Davis Comprehensive Cancer Center, Sacramento, CA; Paul E. Grundy, Stollery Children’s Hospital, University of Alberta, Alberta; Conrad V. Fernandez, IWK Health Center, Dalhousie University Halifax, Nova Scotia, Canada; and Jeffrey S. Dome, Children’s National Health System, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Marcio H. Malogolowkin
- Elizabeth A. Mullen, Dana-Farber Cancer Institute/Boston Children’s Cancer and Blood Disorders Center, Boston, MA; Yueh-Yun Chi and Emily Hibbitts, University of Florida, Gainesville, FL; James R. Anderson, Merck Research Laboratories, North Wales, PA; Katarina J. Steacy, University of Maryland Medical Center, Baltimore, MD; James I. Geller, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Daniel M. Green, St Jude Children’s Research Hospital, Memphis, TN; Geetika Khanna, Washington University School of Medicine, St Louis, MO; Marcio H. Malogolowkin, University of California at Davis Comprehensive Cancer Center, Sacramento, CA; Paul E. Grundy, Stollery Children’s Hospital, University of Alberta, Alberta; Conrad V. Fernandez, IWK Health Center, Dalhousie University Halifax, Nova Scotia, Canada; and Jeffrey S. Dome, Children’s National Health System, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Paul E. Grundy
- Elizabeth A. Mullen, Dana-Farber Cancer Institute/Boston Children’s Cancer and Blood Disorders Center, Boston, MA; Yueh-Yun Chi and Emily Hibbitts, University of Florida, Gainesville, FL; James R. Anderson, Merck Research Laboratories, North Wales, PA; Katarina J. Steacy, University of Maryland Medical Center, Baltimore, MD; James I. Geller, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Daniel M. Green, St Jude Children’s Research Hospital, Memphis, TN; Geetika Khanna, Washington University School of Medicine, St Louis, MO; Marcio H. Malogolowkin, University of California at Davis Comprehensive Cancer Center, Sacramento, CA; Paul E. Grundy, Stollery Children’s Hospital, University of Alberta, Alberta; Conrad V. Fernandez, IWK Health Center, Dalhousie University Halifax, Nova Scotia, Canada; and Jeffrey S. Dome, Children’s National Health System, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Conrad V. Fernandez
- Elizabeth A. Mullen, Dana-Farber Cancer Institute/Boston Children’s Cancer and Blood Disorders Center, Boston, MA; Yueh-Yun Chi and Emily Hibbitts, University of Florida, Gainesville, FL; James R. Anderson, Merck Research Laboratories, North Wales, PA; Katarina J. Steacy, University of Maryland Medical Center, Baltimore, MD; James I. Geller, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Daniel M. Green, St Jude Children’s Research Hospital, Memphis, TN; Geetika Khanna, Washington University School of Medicine, St Louis, MO; Marcio H. Malogolowkin, University of California at Davis Comprehensive Cancer Center, Sacramento, CA; Paul E. Grundy, Stollery Children’s Hospital, University of Alberta, Alberta; Conrad V. Fernandez, IWK Health Center, Dalhousie University Halifax, Nova Scotia, Canada; and Jeffrey S. Dome, Children’s National Health System, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Jeffrey S. Dome
- Elizabeth A. Mullen, Dana-Farber Cancer Institute/Boston Children’s Cancer and Blood Disorders Center, Boston, MA; Yueh-Yun Chi and Emily Hibbitts, University of Florida, Gainesville, FL; James R. Anderson, Merck Research Laboratories, North Wales, PA; Katarina J. Steacy, University of Maryland Medical Center, Baltimore, MD; James I. Geller, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Daniel M. Green, St Jude Children’s Research Hospital, Memphis, TN; Geetika Khanna, Washington University School of Medicine, St Louis, MO; Marcio H. Malogolowkin, University of California at Davis Comprehensive Cancer Center, Sacramento, CA; Paul E. Grundy, Stollery Children’s Hospital, University of Alberta, Alberta; Conrad V. Fernandez, IWK Health Center, Dalhousie University Halifax, Nova Scotia, Canada; and Jeffrey S. Dome, Children’s National Health System, George Washington University School of Medicine and Health Sciences, Washington, DC
| |
Collapse
|
24
|
Wu X, Wang F, Li Y, Wang X, Liu P, Zhang H, Ge Z, Zhang X, Gao C, Chen B. Evaluation of latent membrane protein 1 and microRNA-155 for the prognostic prediction of diffuse large B cell lymphoma. Oncol Lett 2018; 15:9725-9734. [PMID: 29844839 PMCID: PMC5958882 DOI: 10.3892/ol.2018.8560] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 03/16/2018] [Indexed: 12/23/2022] Open
Abstract
Diffuse large B cell lymphoma (DLBCL) has previously been demonstrated to contribute to the mortality of lymphoma with various aggressive features. The prognostic role of the biomarkers latent membrane protein (LMP) 1 and microRNA-(miR)-155 in DLBCL remain controversial. The present study primarily aimed to assess the effect of LMP1 and miR-155 on the survival of DLBCL patients, and additionally evaluate the clinical features to observe their influence on outcomes, compared with previous studies. Formalin-fixed and paraffin-embedded samples were collected from our center between May 2010 and December 2011. Microarray analysis, immunohistochemical analysis and reverse transcription-quantitative polymerase chain reaction were used to evaluate the expression of LMP1 and miR-155. The association between biomarkers or clinical features and patient outcomes was assessed using the log-rank statistical test, Cox proportional hazard model and Kaplan-Meier method. SPSS software was used to statistically analyze the data. A total of 82 patients were included in the present study. The results demonstrated that high expression of LMP1 and miR-155 may be associated with a poor progression-free survival rate, while a high International Prognostic Index score and high expression of LMP1 may be associated with a poor overall survival rate. These results indicated that LMP1 and miR-155 may be novel and reliable biomarkers for the prognostic prediction of lymphoma, and will potentially be analyzed in the future to evaluate patient prognosis.
Collapse
Affiliation(s)
- Xue Wu
- Department of Hematology (Key Department of Jiangsu Medicine), Medical School, The Affiliated Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Fei Wang
- Department of Hematology (Key Department of Jiangsu Medicine), Medical School, The Affiliated Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Yuan Li
- Department of Oncology, Medical School, The Affiliated Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Xiyong Wang
- Department of Oncology, Medical School, The Affiliated Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Ping Liu
- Department of Hematology (Key Department of Jiangsu Medicine), Medical School, The Affiliated Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Haijun Zhang
- Department of Oncology, Medical School, The Affiliated Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Zheng Ge
- Department of Hematology (Key Department of Jiangsu Medicine), Medical School, The Affiliated Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Xiaoping Zhang
- Department of Hematology (Key Department of Jiangsu Medicine), Medical School, The Affiliated Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Chong Gao
- Department of Hematology (Key Department of Jiangsu Medicine), Medical School, The Affiliated Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Baoan Chen
- Department of Hematology (Key Department of Jiangsu Medicine), Medical School, The Affiliated Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
25
|
Mohyuddin GR, Clark AE, Roller J, Shune L, Lin T, Dunavin N, Dias A, Ganguly S, Abhyankar S, McGuirk J, Singh A. Utility of Routine Surveillance Imaging for Hodgkin Disease following Autologous Transplant: Experiences from a Single Institution. Acta Haematol 2018; 139:52-57. [PMID: 29339629 DOI: 10.1159/000484549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 10/24/2017] [Indexed: 11/19/2022]
Abstract
BACKGROUND Surveillance scans performed after autologous stem cell transplant (auto-HCT) for patients with Hodgkin disease (HD) have no proven survival benefit. METHODS We studied survival differences among patients with HD after auto-HCT whose recurrences were detected on clinical history and exam, versus those detected on routine surveillance scan. RESULTS Among the 98 patients with HD that underwent auto-HCT from 2000 to 2014 at our institution, 30 relapsed, of which 21 were detected radiologically and 9 clinically. There were no statistically significant differences in patient characteristics between the 2 groups. The median time to progression was 118 days for the clinical cohort and 284 days for the radiological cohort (p = 0.05). Median overall survival (OS) was 728 days for the clinical cohort, and was not reached for the radiological cohort (p = 0.02). DISCUSSION In our review, most patients with HD after auto-HCT were diagnosed radiologically. Patients whose relapse was diagnosed clinically were likely to be detected earlier and have a shorter OS. Patients with aggressive disease may be detected when clinically relevant, regardless of scanning. Routine scanning may not be necessary in the majority of patients with HD following auto-HCT.
Collapse
Affiliation(s)
- Ghulam Rehman Mohyuddin
- Division of Hematologic Malignancies and Cellular Therapeutic (HMCT), University of Kansas Medical Center, Kansas City, KS, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Tamayo P, Martín A, Díaz L, Cabrero M, García R, García-Talavera P, Caballero D. 18 F-FDG PET/CT in the clinical management of patients with lymphoma. Rev Esp Med Nucl Imagen Mol 2017. [DOI: 10.1016/j.remnie.2017.06.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
27
|
Guidot DM, Switchenko JM, Nastoupil LJ, Koff JL, Blum KA, Maly J, Flowers CR, Cohen JB. Surveillance imaging in mantle cell lymphoma in first remission lacks clinical utility. Leuk Lymphoma 2017; 59:888-895. [PMID: 28797213 DOI: 10.1080/10428194.2017.1361032] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Mantle cell lymphoma (MCL) is a heterogeneous disease with high relapse rates. Limited data guide the use of surveillance imaging following treatment. We constructed a retrospective cohort from two academic institutions of patients with MCL who completed first-line therapy and underwent follow-up for relapse, analyzing the effect of surveillance imaging on survival. Of 217 patients, 102 had documented relapse, with 38 (37%) diagnosed by surveillance imaging and 64 (63%) by other methods. Relapse diagnosis by surveillance imaging had no significant advantage in overall survival from diagnosis date (hazard ratio [HR] = 0.80, p = .39) or relapse date (HR = 0.72, p = .22). Of 801 surveillance images, PET/CT had a positive predictive value (PPV) of 24% and number needed-to-scan/treat (NNT) of 51 to detect one relapse, and CT had a PPV of 49% and NNT of 24. For MCL after first-line therapy, relapse detection by surveillance imaging was not associated with improved survival and lacks clinical benefit.
Collapse
Affiliation(s)
- Daniel M Guidot
- a Winship Cancer Institute, Emory University , Atlanta , GA , USA
| | | | | | - Jean L Koff
- a Winship Cancer Institute, Emory University , Atlanta , GA , USA
| | - Kristie A Blum
- d James Comprehensive Cancer Center, The Ohio State University , Columbus , OH , USA
| | - Joseph Maly
- d James Comprehensive Cancer Center, The Ohio State University , Columbus , OH , USA
| | | | - Jonathon B Cohen
- a Winship Cancer Institute, Emory University , Atlanta , GA , USA
| |
Collapse
|
28
|
Evolution of lymphoma staging and response evaluation: current limitations and future directions. Nat Rev Clin Oncol 2017; 14:631-645. [DOI: 10.1038/nrclinonc.2017.78] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
29
|
Tamayo P, Martín A, Díaz L, Cabrero M, García R, García-Talavera P, Caballero D. 18F-FDG PET/CT in the clinical management of patients with lymphoma. Rev Esp Med Nucl Imagen Mol 2017; 36:312-321. [PMID: 28483374 DOI: 10.1016/j.remn.2017.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/07/2017] [Accepted: 03/13/2017] [Indexed: 01/01/2023]
Abstract
The aim of this work was to review the current recommendations for staging and response assessment of patients with Hodgkin lymphoma (HL) and non-Hodgkin lymphoma (NHL) in routine clinical practice after chemotherapy and/or stem cell transplantation. A five-point scale (5-PS) from the First International Workshop on PET in Lymphoma in Deauville, France, in 2009, was recommended as the standard tool to score imaging to assess treatment response in patients with lymphoma using 18F-Fluorodeoxyglucose (FDG) PET/CT. Following the recommendations of the 11th and 12th International Conferences on Malignant Lymphoma held in Lugano (Switzerland), in 2011 and 2013, respectively, a consensus (the so-called Lugano Classification) was reached regarding the use of PET/CT for staging and response assessment in FDG-avid lymphomas. As a result, 18F-FDG PET/CT was formally incorporated into standard staging for FDG-avid lymphomas. A bone marrow biopsy is no longer indicated for the routine staging of HL and most diffuse large B-cell lymphomas. PET/CT will be used to assess response in FDG-avid histologies using the 5-point scale. The recent introduction of biological agents with immune mechanisms requires flexibility in interpretations of the Lugano criteria due to tumour flare or a pseudo-progression effect produced by these agents. Provisional criteria have been proposed (Lymphoma Response to Immunomodulatory Therapy Criteria) with the introduction of the term 'Indeterminate Response' in order to identify this phenomenon until confirmed as flare/pseudoprogression or true progression. All these recommendations will improve evaluations of patients with lymphoma, and allow comparison of results from clinical practice and trials.
Collapse
Affiliation(s)
- P Tamayo
- Servicio de Medicina Nuclear, Hospital Universitario de Salamanca, Salamanca, España; Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, España.
| | - A Martín
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, España; Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, España
| | - L Díaz
- Servicio de Medicina Nuclear, Hospital Universitario de Salamanca, Salamanca, España
| | - M Cabrero
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, España; Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, España
| | - R García
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, España; Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, España
| | - P García-Talavera
- Servicio de Medicina Nuclear, Hospital Universitario de Salamanca, Salamanca, España
| | - D Caballero
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, España; Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, España
| |
Collapse
|