188 results on '"Scott A. Waldman"'
Search Results
2. Guanylyl cyclase 2C (GUCY2C) in gastrointestinal cancers: recent innovations and therapeutic potential
- Author
-
Scott A. Waldman, Adam E. Snook, and Ariana A Entezari
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Clinical Biochemistry ,Receptors, Enterotoxin ,Article ,03 medical and health sciences ,0302 clinical medicine ,Gastrointestinal Agents ,Drug Discovery ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,Guanylyl Cyclase 2C ,Gastrointestinal Neoplasms ,Pharmacology ,business.industry ,Immunotherapy ,Guanylate cyclase 2C ,Signaling system ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Molecular Medicine ,business ,Signal Transduction - Abstract
INTRODUCTION: Gastrointestinal (GI) cancers account for the second leading cause of cancer-related deaths in the United States. Guanylyl cyclase C (GUCY2C) is an intestinal signaling system that regulates intestinal fluid and electrolyte secretion as well as intestinal homeostasis. In recent years, it has emerged as a promising target for chemoprevention and therapy for GI malignancies. AREAS COVERED: The loss of GUCY2C signaling early in colorectal tumorigenesis suggests it could have a significant impact on tumor initiation. Recent studies highlight the importance of GUCY2C signaling in preventing colorectal tumorigenesis using agents such as linaclotide, plecanatide and sildenafil. Further, GUCY2C is a novel target for immunotherapy and a diagnostic marker of primary and metastatic disease. EXPERT OPINION: There is an unmet need for prevention and therapy in GI cancers. In that context, GUCY2C is a promising target for prevention, although the precise mechanisms by which GUCY2C signaling affects tumorigenesis remain to be defined. Further, clinical trials are exploring its role as an immunotherapeutic target for vaccines to prevent metastatic disease. Indeed, GUCY2C is an emerging target across the disease continuum from chemoprevention, to diagnostic management, through the treatment and prevention of metastatic disease.
- Published
- 2021
3. Review article: diagnosis, management and patient perspectives of the spectrum of constipation disorders
- Author
-
Scott A. Waldman, Kimberly D. Orleck, Kimberly Kearns, Amol Sharma, and Satish S.C. Rao
- Subjects
medicine.medical_specialty ,Chronic constipation ,Constipation ,Hepatology ,business.industry ,medicine.medical_treatment ,Anorectal manometry ,Gastroenterology ,MEDLINE ,Review Article ,Biofeedback ,Review article ,Chronic idiopathic constipation ,medicine ,Defecation ,Pharmacology (medical) ,medicine.symptom ,Intensive care medicine ,business ,Review Articles - Abstract
Summary Background Chronic constipation is a common, heterogeneous disorder with multiple symptoms and pathophysiological mechanisms. Patients are often referred to a gastroenterology provider after laxatives fail. However, there is limited knowledge of the spectrum and management of constipation disorders. Aim To discuss the latest understanding of the spectrum of constipation disorders, tools for identifying a pathophysiologic‐based diagnosis in the specialist setting, treatment options and the patient's perspective of constipation. Methods Literature searches were conducted using PubMed for constipation diagnostic criteria, diagnostic tools and approved treatments. The authors provided insight from their own practices. Results Clinical assessment, stool diaries and Rome IV diagnostic criteria can facilitate diagnosis, evaluate severity and distinguish between IBS with constipation, chronic idiopathic constipation and dyssynergic defecation. Novel smartphone applications can help track constipation symptoms. Rectal examinations, anorectal manometry and balloon expulsion, assessments of neuromuscular function with colonic transit time and colonic manometry can provide mechanistic understanding of underlying pathophysiology. Treatments include lifestyle and diet changes, biofeedback therapy and pharmacological agents. Several classes of laxatives, as well as prokinetic and prosecretory agents, are available; here we describe their mechanisms of action, efficacy and side effects. Conclusions Constipation includes multiple overlapping subtypes identifiable using detailed history, current diagnostic tools and smartphone applications. Recognition of individual subtype(s) could pave the way for optimal, evidence‐based treatments by a gastroenterology provider., Gastroenterology providers should be familiar with the constipation subtypes and their underlying pathophysiology, as well as key signalling pathways that may contribute to constipation. Familiarisation with the varied mechanisms of action, expected efficacy and side effects of available therapies helps to select the appropriate treatment and meet patient expectations.
- Published
- 2021
4. Phase I double-blind, placebo-controlled trial of dolcanatide (SP-333) 27 mg to explore colorectal bioactivity in healthy volunteers
- Author
-
David Kastenberg, Ryan McMurray, Christopher H. Henry, Walter K. Kraft, Paul J. Limburg, David S. Weinberg, Leo Katz, Scott A. Waldman, Nathan R. Foster, Gary Della'Zanna, Angela Pallotto, and Stephanie Moleski
- Subjects
Agonist ,Cancer Research ,medicine.drug_class ,Guanylin ,Receptors, Enterotoxin ,Pharmacology ,chemistry.chemical_compound ,Double-Blind Method ,medicine ,Humans ,Large intestine ,Receptor ,Linaclotide ,Cyclic GMP ,business.industry ,Guanylate cyclase 2C ,Healthy Volunteers ,medicine.anatomical_structure ,Oncology ,chemistry ,Receptors, Guanylate Cyclase-Coupled ,Molecular Medicine ,business ,Colorectal Neoplasms ,Peptides ,Uroguanylin ,Hormone ,Research Paper - Abstract
Guanylyl cyclase C (GUCY2C) is a tumor-suppressing receptor silenced by loss of expression of the luminocrine hormones guanylin and uroguanylin early in colorectal carcinogenesis. This observation suggests oral replacement with a GUCY2C agonist may be an effective targeted chemoprevention agent. Previous studies revealed that linaclotide, an oral GUCY2C agonist formulated for gastric release, did not persist to activate guanylyl cyclase signaling in the distal rectum. Dolcanatide is an investigational oral uroguanylin analog, substituted with select D amino acids, for enhanced stability and extended persistence to activate GUCY2C in small and large intestine. However, the ability of oral dolcanatide to induce a pharmacodynamic (PD) response by activating GUCY2C in epithelial cells of the colorectum in humans remains undefined. Here, we demonstrate that administration of oral dolcanatide 27 mg daily for 7 d to healthy volunteers did not activate GUCY2C, quantified as accumulation of its product cyclic GMP, in epithelial cells of the distal rectum. These data reveal that the enhanced stability of dolcanatide, with persistence along the rostral-caudal axis of the small and large intestine, is inadequate to regulate GUCY2C across the colorectum to prevent tumorigenesis. These results highlight the importance of developing a GUCY2C agonist for cancer prevention formulated for release and activity targeted to the colorectum.
- Published
- 2021
5. Biomarker targeting of colorectal cancer stem cells
- Author
-
Scott A. Waldman, Adam E. Snook, Ellen M. Caparosa, and Jonathan Stem
- Subjects
business.industry ,Colorectal cancer ,medicine.medical_treatment ,Biochemistry (medical) ,Clinical Biochemistry ,Antineoplastic Agents ,Immunotherapy ,medicine.disease ,Cancer stem cell ,Drug Discovery ,Biomarkers, Tumor ,Neoplastic Stem Cells ,Cancer research ,Humans ,Medicine ,Biomarker (medicine) ,Molecular Targeted Therapy ,Stem cell ,Colorectal Neoplasms ,business - Published
- 2019
6. Silencing the GUCA2A-GUCY2C tumor suppressor axis in CIN, serrated, and MSI colorectal neoplasia
- Author
-
Ying Feng, Babar Bashir, Juan P. Palazzo, Scott A. Waldman, Eric R. Fearon, Dante J. Merlino, Adam E. Snook, Jeffrey A. Rappaport, and Esteban Gnass
- Subjects
Adenoma ,Adult ,Male ,0301 basic medicine ,Colorectal cancer ,Receptors, Enterotoxin ,Mice, Transgenic ,Article ,Pathology and Forensic Medicine ,law.invention ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Tubular adenoma ,law ,medicine ,Animals ,Humans ,Gene silencing ,Receptor ,CDX2 ,neoplasms ,Aged ,business.industry ,Microsatellite instability ,Middle Aged ,medicine.disease ,digestive system diseases ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Suppressor ,Female ,DNA mismatch repair ,Colorectal Neoplasms ,business ,Signal Transduction - Abstract
Colorectal cancers (CRCs) initiate through distinct mutations, including in APC pathway components leading to tubular adenomas (TAs); in BRAF, with epigenetic silencing of CDX2, leading to serrated adenomas (SAs); and in the DNA mismatch repair machinery driving microsatellite instability (MSI). Transformation through the APC pathway involves loss of the hormone GUCA2A that silences the tumor-suppressing receptor GUCY2C. Indeed, oral hormone replacement is an emerging strategy to reactivate GUCY2C and prevent CRC initiation and progression. Moreover, retained expression by tumors arising from TAs has established GUCY2C as a diagnostic and therapeutic target to prevent and treat metastatic CRC. Here, we defined the potential role of the GUCA2A-GUCY2C axis and its suitability as a target in tumors arising through the SA and MSI pathways. GUCA2A hormone expression was eliminated in TAs, SAs, and MSI tumors compared to their corresponding normal adjacent tissues. In contrast to the hormone, the tumor-suppressing receptor GUCY2C was retained in TA and MSI tumors. Surprisingly, GUCY2C expression was nearly eliminated in SAs, reflecting loss of the transcription factor CDX2. Changes in the GUCA2A-GUCY2C axis in human SAs and MSI tumors were precisely recapitulated in genetic mouse models. These data reveal the possibility of GUCA2A loss silencing GUCY2C in the pathophysiology of, and oral hormone replacement to restore GUCY2C signaling to prevent, MSI tumors. Also, they highlight the potential for targeting GUCY2C to prevent and treat metastases arising from TA and MSI tumors. In contrast, loss of GUCY2C excludes patients with SAs as candidates for GUCY2C-based prevention and therapy.
- Published
- 2019
7. Therapeutic targeting of gastrointestinal cancer stem cells
- Author
-
Ellen M. Caparosa, John C. Flickinger, Scott A. Waldman, Adam E. Snook, Jonathan Stem, and Dante J. Merlino
- Subjects
Embryology ,medicine.medical_treatment ,0206 medical engineering ,Biomedical Engineering ,Review ,02 engineering and technology ,Disease ,Metastasis ,03 medical and health sciences ,Cancer stem cell ,Humans ,Medicine ,Molecular Targeted Therapy ,Gastrointestinal cancer ,Gastrointestinal Neoplasms ,030304 developmental biology ,0303 health sciences ,business.industry ,Cancer ,Immunotherapy ,medicine.disease ,020601 biomedical engineering ,Cancer cell ,Neoplastic Stem Cells ,Cancer research ,Stem cell ,business ,Oxidation-Reduction ,DNA Damage - Abstract
Gastrointestinal cancers remain a tremendous burden on society. Despite advances in therapy options, including chemotherapy and radiation, cancer mortality from recurrences and metastases occur frequently. Cancer stem cells (CSCs) drive disease recurrence and metastasis, as these cells are uniquely equipped to self-renew and evade therapy. Therefore, cancer eradication requires treatment strategies that target CSCs in addition to differentiated cancer cells. This review highlights current literature on therapies targeting CSCs in gastrointestinal cancer.
- Published
- 2019
8. Health Care Evolves From Reactive to Proactive
- Author
-
Scott A. Waldman and Andre Terzic
- Subjects
Pharmacology ,Discovery science ,Organizational innovation ,Extramural ,business.industry ,Psychological intervention ,MEDLINE ,Disease ,030226 pharmacology & pharmacy ,Article ,Organizational Innovation ,Primary Prevention ,03 medical and health sciences ,0302 clinical medicine ,Nursing ,030220 oncology & carcinogenesis ,Intervention (counseling) ,Health care ,Humans ,Pharmacology (medical) ,Psychology ,business ,Delivery of Health Care - Abstract
Decoding health and disease pathways drives healthcare evolution. Historically, therapeutic paradigms have relied on interventions that mitigate symptoms of established diseases. Increasingly, molecular insights into pathophysiology now provide unprecedented opportunities to offer curative solutions or even prevent disease and thereby secure longitudinal wellness. These opportunities extend past individual patients to entire populations and geographies. Moreover, they optimize prospective healthspan across lifespan. Linking discovery science and its translatable innovations beyond reactive disease intervention to proactive prevention will maximize society’s returns creating the greatest benefit for the greatest number of people globally.
- Published
- 2018
9. GUCY2C as a biomarker to target precision therapies for patients with colorectal cancer
- Author
-
Megan Weindorfer, Amanda Lisby, Babar Bashir, Sanjna Shelukar, Adam E. Snook, John C. Flickinger, Scott A. Waldman, and Madison Crutcher
- Subjects
Pharmacology ,Oncology ,medicine.medical_specialty ,Colorectal cancer ,business.industry ,Disease ,Guanylate cyclase 2C ,medicine.disease ,digestive system diseases ,Article ,Internal medicine ,Drug Discovery ,Genetics ,medicine ,Molecular Medicine ,CAR T-cell therapy ,Biomarker (medicine) ,Cancer vaccine ,Stage (cooking) ,Antibody therapy ,business ,neoplasms - Abstract
INTRODUCTION: Colorectal cancer (CRC) is one of the most-deadly malignancies worldwide. Current therapeutic regimens for CRC patients are relatively generic, based primarily on disease type and stage, with little variation. As the field of molecular oncology advances, so too must therapeutic management of CRC. Understanding molecular heterogeneity has led to a new-found promotion for precision therapy in CRC; underlining the diversity of molecularly targeted therapies based on individual tumor characteristics. AREAS COVERED: We review current approaches for the treatment of CRC and discuss the potential of precision therapy in advanced CRC. We highlight the utility of the intestinal protein guanylyl cyclase C (GUCY2C), as a multi-purpose biomarker and unique therapeutic target in CRC. Here, we summarize current GUCY2C-targeted approaches for treatment of CRC. EXPERT OPINION: The GUCY2C biomarker has multi-faceted utility in medicine. Developmental investment of GUCY2C as a diagnostic and therapeutic biomarker offers a variety of options taking the molecular characteristics of cancer into account. From GUCY2C-targeted therapies, namely cancer vaccines, CAR-T cells, and monoclonal antibodies, to GUCY2C agonists for chemoprevention in those who are at high risk for developing colorectal cancer, the utility of this protein provides many avenues for exploration with significance in the field of precision medicine.
- Published
- 2021
10. Emerging targets for the diagnosis of Parkinson's disease: examination of systemic biomarkers
- Author
-
Scott A. Waldman, Adam E. Snook, and Lara Cheslow
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Parkinson's disease ,Clinical Biochemistry ,Context (language use) ,Disease ,Review ,Unmet needs ,03 medical and health sciences ,0302 clinical medicine ,Drug Discovery ,medicine ,Animals ,Humans ,Intensive care medicine ,Organ system ,business.industry ,Biochemistry (medical) ,Advanced stage ,Neurodegeneration ,Disease progression ,Parkinson Disease ,medicine.disease ,030104 developmental biology ,Disease Progression ,business ,030217 neurology & neurosurgery ,Biomarkers - Abstract
Parkinson’s disease (PD) is a highly prevalent and irreversible neurodegenerative disorder that is typically diagnosed in an advanced stage. Currently, there are no approved biomarkers that reliably identify PD patients before they have undergone extensive neuronal damage, eliminating the opportunity for future disease-modifying therapies to intervene in disease progression. This unmet need for diagnostic and therapeutic biomarkers has fueled PD research for decades, but these efforts have not yet yielded actionable results. Recently, studies exploring mechanisms underlying PD progression have offered insights into multisystemic contributions to pathology, challenging the classic perspective of PD as a disease isolated to the brain. This shift in understanding has opened the door to potential new biomarkers from multiple sites in the body. This review focuses on emerging candidates for PD biomarkers in the context of current diagnostic approaches and multiple organ systems that contribute to disease.
- Published
- 2021
11. Stem cells as therapeutic targets in colorectal cancer
- Author
-
Adam E. Snook, Alicja Zalewski, and Scott A. Waldman
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Colorectal cancer ,medicine.medical_treatment ,Disease ,Immunotherapy, Adoptive ,Article ,Metastasis ,03 medical and health sciences ,Therapeutic approach ,0302 clinical medicine ,Cancer stem cell ,Internal medicine ,medicine ,Humans ,Hedgehog Proteins ,AC133 Antigen ,Immune Checkpoint Inhibitors ,beta Catenin ,Janus Kinases ,Pharmacology ,Receptors, Notch ,business.industry ,Stem Cells ,Cancer ,General Medicine ,Immunotherapy ,medicine.disease ,Wnt Proteins ,MicroRNAs ,STAT Transcription Factors ,030104 developmental biology ,030220 oncology & carcinogenesis ,Molecular Medicine ,RNA, Long Noncoding ,Stem cell ,business ,Colorectal Neoplasms ,Signal Transduction - Abstract
Colorectal cancer continues to represent a significant burden on public health as the second highest cause of cancer mortality, when men and women are combined, in the US. About 50% of patients either present with late-stage metastatic disease, or develop metastatic recurrences, and ultimately die. In turn, this mortality largely reflects cancer stem cells, tumor-initiating cells that are responsible for cancer progression, drug resistance, recurrence and metastasis. This review summarizes the unique properties of colorectal cancer stem cells, and the emerging strategies by which they can be selectively targeted as a therapeutic approach to eradicating this disease.
- Published
- 2021
12. From leptin to lasers: the past and present of mouse models of obesity
- Author
-
Scott A. Waldman, Adam E. Snook, and Joshua R Barton
- Subjects
Leptin ,media_common.quotation_subject ,Gut–brain axis ,Appetite ,Bioinformatics ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Drug Discovery ,Brain-Gut Axis ,medicine ,Animals ,Leptin resistance ,Obesity ,030304 developmental biology ,media_common ,0303 health sciences ,business.industry ,Lasers ,medicine.disease ,Syndromic obesity ,Neuromodulation (medicine) ,030220 oncology & carcinogenesis ,business - Abstract
INTRODUCTION. Obesity is a prevalent condition that accounts for significant morbidity and mortality across the globe. Despite substantial effort, most obesity pharmacotherapies have proven unsafe or ineffective. The use of obese mouse models provides unique insight into the hormones and mechanisms that regulate appetite and metabolism. Paramount among these models are the “obese” and “diabetic” mice that revealed the powerful satiety hormone leptin, revolutionizing obesity research. AREAS COVERED. In this article, we discuss work on leptin therapy, and the clinical response to leptin in humans. The authors describe the use of modern mouse genetics to study targetable mechanisms for genetic forms of human obesity. Additionally, they describe mouse models of neuromodulation and their utility in unraveling neural circuits that govern appetite and metabolism. EXPERT OPINION. Combining past and present models of obesity is required for the development of safe, effective, and impactful obesity therapy. Current research in obesity can benefit from repositories of genetically engineered mouse models to discover interactions between appetitive systems and circuits. Combining leptin therapy with other satiety signals comprising the gut-brain axis is a promising approach to induce significant enduring weight loss.
- Published
- 2021
13. Guanylyl cyclase C as a biomarker for immunotherapies for the treatment of gastrointestinal malignancies
- Author
-
Joshua R Barton, John C. Flickinger, Jeffrey A. Rappaport, Trevor R. Baybutt, Amanda M. Pattison, Adam E. Snook, and Scott A. Waldman
- Subjects
0301 basic medicine ,Colorectal cancer ,medicine.medical_treatment ,Clinical Biochemistry ,Receptors, Enterotoxin ,Context (language use) ,Review ,03 medical and health sciences ,0302 clinical medicine ,Drug Discovery ,medicine ,Humans ,Gastrointestinal cancer ,Molecular Targeted Therapy ,Gastrointestinal Neoplasms ,business.industry ,Stomach ,Biochemistry (medical) ,Cancer ,Immunotherapy ,Guanylate cyclase 2C ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Pancreas ,business ,Biomarkers - Abstract
Gastrointestinal cancers encompass a diverse class of tumors arising in the GI tract, including esophagus, stomach, pancreas and colorectum. Collectively, gastrointestinal cancers compose a high fraction of all cancer deaths, highlighting an unmet need for novel and effective therapies. In this context, the transmembrane receptor guanylyl cyclase C (GUCY2C) has emerged as an attractive target for the prevention, detection and treatment of many gastrointestinal tumors. GUCY2C is an intestinally-restricted protein implicated in tumorigenesis that is universally expressed by primary and metastatic colorectal tumors as well as ectopically expressed by esophageal, gastric and pancreatic cancers. This review summarizes the current state of GUCY2C-targeted modalities in the management of gastrointestinal malignancies, with special focus on colorectal cancer, the most incident gastrointestinal malignancy.
- Published
- 2021
14. 739 Development of patient-derived models of esophageal cancer for GUCY2C-directed immunotherapeutic testing
- Author
-
David E. Loren, Alicja Zalewski, Adam E. Snook, Scott A. Waldman, Megan Weindorfer, Robert D Carlson, Nathaniel R. Evans, Madison Crutcher, Trevor R. Baybutt, Ernest L. Rosato, and Amanda Lisby
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Colorectal cancer ,medicine.medical_treatment ,Cancer ,Esophageal cancer ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Primary tumor ,lcsh:RC254-282 ,Esophagectomy ,Internal medicine ,medicine ,Adenocarcinoma ,business ,Survival rate ,Neoadjuvant therapy - Abstract
Background Esophageal cancer is the fifth most common cause of cancer-related death in the world1 with a 5-year survival rate of Methods To develop esophageal cancer models for GUCY2C immunotherapy testing, esophageal cancer samples were collected at Thomas Jefferson University Hospital by endoscopic biopsy of treatment-naive patients or by esophagectomy, primarily in patients previously treated with standard neoadjuvant therapy. Patient-derived xenograft (PDX) models were initiated from samples to establish in vivo models for immunotherapy testing. qRT-PCR, immunoblot, and immunofluorescence were performed to test for GUCY2C expression in primary and PDX specimens. Histopathology was performed to confirm retention of primary tumor features. Results GUCY2C was present in only 2 of 6 esophagectomy samples. Interestingly, those patients with detectable GUCY2C were treatment-naive, while neoadjuvant-treated patients lacked viable tumor, revealing neoadjuvant therapy as a significant barrier to esophageal cancer model generation. In contrast, of the 3 adenocarcinoma specimens collected by endoscopic biopsy in treatment-naive patients, 2 express GUCY2C. To date, PDX models have been initiated from 6 total samples and successfully established for 3 samples. This 50% success rate may improve over time as PDX formation is often delayed in many models (>150 days). Importantly, established esophageal adenocarcinoma PDX models were histologically similar to their matched primary tumors and retained GUCY2C expression, integral to their validation as models of GUCY2C immunotherapy testing. Conclusions Several human esophageal adenocarcinoma models were successfully established, primarily from endoscopic biopsy of treatment-naive patients as neoadjuvant therapy proved to be a significant barrier. These models will be useful to explore GUCY2C-directed CAR-T cell therapies and other novel therapies targeting intestine-like esophageal cancer, prior to testing in early-phase clinical trials. Acknowledgements The authors thank the Translational Research & Pathology Core Facility and the Office of Animal Resources at Thomas Jefferson University for their continued support to make this research possible. The authors would also like to thank the Clinical Research Unit at Thomas Jefferson University for their assistance in the collection of patient specimens. This work was supported by a DeGregorio Family Foundation Award and by the Department of Defense Congressionally Directed Medical Research Programs (W81XWH-17-1-0299, W81XWH-191-0263, and W81XWH-19-1-0067) to AES. SAW is supported by the National Institutes of Health (NIH) (R01 CA204881, R01 CA206026, and P30 CA56036), the Defense Congressionally Directed Medical Research Program W81XWH-17-PRCRP-TTSA, and Targeted Diagnostic & Therapeutics. SAW and AES were also supported by a grant from The Courtney Ann Diacont Memorial Foundation. SAW is the Samuel M.V. Hamilton Professor of Thomas Jefferson University. AZ and MC were supported by NIH institutional award T32 GM008562 for Postdoctoral Training in Clinical Pharmacology. Ethics Approval The study was approved by the Thomas Jefferson University Institutional Review Board (#18D.495) and Institutional Animal Care and use Committee (#01529). References Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin2018;68:394–424. doi:10.3322/caac.21492. Yousefi MS, Sharifi-Esfahani M, Pourgholam-Amiji N, Afshar M, Sadeghi-Gandomani H, Otroshi O, et al. Esophageal cancer in the world: incidence, mortality and risk factors. Biomed Res Ther 2018;5:2504–17. doi:10.15419/bmrat.v5i7.460. Huang F-L, Yu S-J. Esophageal cancer: risk factors, genetic association, and treatment. Asian J Surg 2018;41:210–5. doi:10.1016/j.asjsur.2016.10.005. Magee MS, Abraham TS, Baybutt TR, Flickinger JC, Ridge NA, Marszalowicz GP, et al. Human GUCY2C-targeted chimeric antigen receptor (CAR)-expressing T cells eliminate colorectal cancer metastases. Cancer Immunol Res 2018;6:509–16. doi:10.1158/2326-6066.CIR-16-0362. Magee MS, Kraft CL, Abraham TS, Baybutt TR, Marszalowicz GP, Li P, et al. GUCY2C-directed CAR-T cells oppose colorectal cancer metastases without autoimmunity. Oncoimmunology 2016;5:e1227897. doi:10.1080/2162402X.2016.1227897.
- Published
- 2020
15. 105 A third-generation human GUCY2C-targeted CAR-T cell for colorectal cancer immunotherapy
- Author
-
Trevor R. Baybutt, Adam E. Snook, Jonathan Stem, Ellen M. Caparosa, Alicja Zalewski, and Scott A. Waldman
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,education.field_of_study ,Magnetic-activated cell sorting ,business.industry ,medicine.medical_treatment ,Population ,Cancer ,Immunotherapy ,medicine.disease ,Chimeric antigen receptor ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Cell killing ,030220 oncology & carcinogenesis ,Internal medicine ,Cancer cell ,medicine ,Nivolumab ,business ,education - Abstract
Background Colorectal cancer (CRC) presents a significant public health burden, responsible for the second most cancer-related deaths in the United States, with an increasing incidence in young adults observed globally.1,2 While the blockade of immune checkpoints received FDA approval as a CRC therapeutic, only patients with microsatellite instability, accounting for 15% of sporadic cases, demonstrate partial or complete responses.3 We present a third-generation chimeric antigen receptor (CAR)-T cell directed towards the extracellular domain of the mucosal antigen guanylyl cyclase C (GUCY2C), which is over-expressed in 80% of CRC cases, as a therapeutic alternative for late stage disease. Here, we demonstrate that human GUCY2C CAR-T cells can selectively kill GUCY2C-expressing colorectal cancer cells in vitro and produce inflammatory cytokines in response to antigenic stimulation. Methods Peripheral blood mononuclear (PBMCs) cells were isolated from leukoreduction filters obtained from the Thomas Jefferson University Hospital Blood Donor Center (IRB #18D.495). Magnetic Activated Cell Sorting (MACS) technology was used to negatively select pan-T cells (Miltenyi Biotec), followed by activation and expansion using anti-CD3, anti-CD28, and anti-CD2 coated microbeads (Miltenyi Biotec) and supplemented with IL-7 and IL-15 (Biological Resources Branch Preclinical Biologics Repository – NCI). T-cells were transduced with a lentiviral vector encoding the anti-GUCY2C CAR. Our CAR utilizes a single chain variable fragment of human origin directed towards the extracellular domain of GUCY2C, the CD28 hinge, transmembrane, and intracellular signaling domain (ICD), 4-1BB (CD137) ICD, and CD3ζ ICD. CAR-T cells were used for experiments between 10 to 14 days after activation in vitro using the xCELLigence real time cytotoxicity assay and intracellular cytokine staining. Results GUCY2C-directed CAR-T cells specifically lysed the GUCY2C-expressing metastatic CRC cell line T84, while the control CAR did not. GUCY2C-negative CRC cells were not killed by either. In addition to cell killing, GUCY2C-directed CAR-T cells of both the CD8+ and CD4+ co-receptor lineage produced the inflammatory cytokines IFN-γ and TNFα in response to GUCY2C antigen. Conclusions We demonstrate that human GUCY2C-directed CAR-T cells can selectively target GUCY2C-expressing cancer cells. We hypothesize that GUCY2C-directed CAR-T cells present a viable therapeutic option for metastatic CRC. In vivo animal models to examine this potential are currently on-going. Acknowledgements This work was supported by the Department of Defense Congressionally Directed Medical Research Programs (W81XWH-17-1-0299, W81XWH-191-0263, and W81XWH-19-1-0067) to AES and Targeted Diagnostic & Therapeutics to SAW. AES is also supported by a DeGregorio Family Foundation Award. SAW is supported by the National Institutes of Health (NIH) (R01 CA204881, R01 CA206026, and P30 CA56036), and the Department of Defense Congressionally Directed Medical Research Program W81XWH-17-PRCRP-TTSA. SAW and AES were also supported by a grant from The Courtney Ann Diacont Memorial Foundation. SAW is the Samuel M.V. Hamilton Professor of Thomas Jefferson University. JS, EC, and AZ were supported by an NIH institutional award T32 GM008562 for Postdoctoral Training in Clinical Pharmacology. Ethics Approval This study was approved by the Thomas Jefferson University Institutional Review Board (IRB Control #18D.495) and the Institutional Animal Care and Use Committee (Protocol #01529). References Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin2020;70: 7–30. doi:10.3322/caac.21590 Araghi M, Soerjomataram I, Bardot A, Ferlay J, Cabasag CJ, Morrison DS, et al. Changes in colorectal cancer incidence in seven high-income countries: a population-based study. Lancet Gastroenterol Hepatol 2019;4: 511–518. doi:10.1016/S2468-1253(19)30147-5 Overman MJ, McDermott R, Leach JL, Lonardi S, Lenz H-J, Morse MA, et al. Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study. Lancet Oncol 2017;18: 1182–1191. doi:10.1016/S1470-2045(17)30422-9
- Published
- 2020
16. 122 Guanylyl cyclase C as a target for CAR-T cell therapy in a metastatic gastric cancer model
- Author
-
Trevor R. Baybutt, Adam E. Snook, Megan Weindorfer, Amanda Lisby, Scott A. Waldman, Alicja Zalewski, and Robert D Carlson
- Subjects
Colorectal cancer ,business.industry ,Cancer ,Guanylate cyclase 2C ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,Chimeric antigen receptor ,Cell therapy ,Antigen ,Cancer research ,medicine ,Adenocarcinoma ,Enterocyte differentiation ,business - Abstract
Background Gastric cancer is the sixth most common cancer and second-leading cause of cancer-related mortality worldwide.1 The heterogenous and genetically complex nature of this disease underlies the challenges in developing effective therapies for metastatic gastric cancer. In the majority of cases, stomach tumors evolve from intestinal metaplasia resulting in ectopic expression of the enterocyte differentiation antigen guanylyl cyclase C (GUCY2C) by ~50% of primary and metastatic gastric cancers.2–4 In the context of the efficacy of GUCY2C-directed chimeric antigen receptor (CAR)-T cells against metastatic colorectal cancer in animal models,5,6 we hypothesized that this adoptive cell therapy may be effective against metastatic gastric cancer. Methods Here, we explored the efficacy of GUCY2C-directed CAR-T cells for gastric cancer in a patient derived xenograft (PDX) tumor model. Also, we interrogated translational GUCY2C biomarker assays using RT-qPCR, immunoblot analysis, and immunohistochemistry (IHC) for the intended purpose of identifying patients whose tumors express GUCY2C and could benefit from GUCY2C-directed CAR-T cell therapy. Results GUCY2C-directed CAR-T cells significantly reduced subcutaneous T84 colorectal tumor growth, producing a 5-fold reduction in tumor volume, compared to control treated tumors. GUCY2C-directed CAR-T cells produced no response in tumors produced from the GUCY2C-deficient colorectal cancer cell line, SW480. Importantly, GUCY2C-directed CAR-T cells controlled gastric cancer PDX growth, maintaining a >12-fold reduction in tumor volume compared to control and in some cases produced complete tumor elimination. Furthermore, IHC based assays, indicate that antibodies developed in our laboratory may be suitable for development of a companion diagnostic for GUCY2C-directed CAR-T cells. Indeed, the commercial polyclonal antibody demonstrated robust, non-specific staining regardless of tissue type or GUCY2C mRNA profile, while novel monoclonal antibodies produced in our laboratory primarily detected protein localized to the membrane of glandular epithelial cells, demonstrating antigen specificity, and indicating their potential for further development in diagnostic companion assays to identify gastric cancer patients who may benefit from GUCY2C-directed CAR-T cell therapy. Conclusions GUCY2C-directed CAR-T cells prevented the growth of, and at times eliminated, a subcutaneous gastric cancer PDX model. In the context of previously established safety in mouse models, additional studies defining the efficacy of GUCY2C-directed CAR-T cells in gastric cancer models may allow future translation of this therapy to patients with advanced gastric cancers. Concurrent development of a novel companion diagnostic IHC assay would permit identification of the ~50% of gastric cancer patients whose tumors express GUCY2C and could benefit from this therapy. Acknowledgements This work was supported by a DeGregorio Family Foundation Award and by the Department of Defense Congressionally Directed Medical Research Programs (W81XWH-17-1-0299, W81XWH-191-0263, and W81XWH-19-1-0067) to AES. SAW is supported by the National Institutes of Health (NIH) (R01 CA204881, R01 CA206026, and P30 CA56036), the Defense Congressionally Directed Medical Research Program W81XWH-17-PRCRP-TTSA, and Targeted Diagnostic & Therapeutics. SAW and AES were also supported by a grant from The Courtney Ann Diacont Memorial Foundation. SAW is the Samuel M.V. Hamilton Professor of Thomas Jefferson University. AZ was supported by NIH institutional award T32 GM008562 for Postdoctoral Training in Clinical Pharmacology.The authors thank the NCI Patient-Derived Models Repository for their support and resources to make this research possible. The authors also thank the Sidney Kimmel Cancer Center Translational Research & Pathology Core Facility, and the Office of Animal Resources at Thomas Jefferson University for their continued technical assistance and support in this research. Ethics Approval This study was approved by the Thomas Jefferson University Institutional Review Board (#14.0204) and the Instituational Animal Care and Use Commitee (Protocol #01529). References Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin2018;68:394–424. doi:10.3322/caac.21492. Park J, Schulz S, Haaf J, Kairys JC, Waldman SA. Ectopic expression of guanylyl cyclase C in adenocarcinomas of the esophagus and stomach. Cancer Epidemiol Biomarkers Prev 2002;11:739–44. Birbe R, Palazzo JP, Walters R, Weinberg D, Schulz S, Waldman SA. Guanylyl cyclase C is a marker of intestinal metaplasia, dysplasia, and adenocarcinoma of the gastrointestinal tract. Hum Pathol. 2005;36:170–9. doi:10.1016/j.humpath.2004.12.002. Mathur D, Root AR, Bugaj-Gaweda B, Bisulco S, Tan X, Fang W, et al. A Novel GUCY2C-CD3 T-Cell Engaging Bispecific Construct (PF-07062119) for the Treatment of Gastrointestinal Cancers. Clin Cancer Res 2020;26:2188–202. doi:10.1158/1078-0432.CCR-19-3275. Magee MS, Kraft CL, Abraham TS, Baybutt TR, Marszalowicz GP, Li P, et al. GUCY2C-directed CAR-T cells oppose colorectal cancer metastases without autoimmunity. Oncoimmunology 2016;5:e1227897. doi:10.1080/2162402X.2016.1227897. Magee MS, Abraham TS, Baybutt TR, Flickinger JC, Ridge NA, Marszalowicz GP, et al. Human GUCY2C-Targeted Chimeric Antigen Receptor (CAR)-Expressing T Cells Eliminate Colorectal Cancer Metastases. Cancer Immunol Res 2018;6:509–16. doi:10.1158/2326-6066.CIR-16-0362.
- Published
- 2020
17. An update on guanylyl cyclase C in the diagnosis, chemoprevention, and treatment of colorectal cancer
- Author
-
Scott A. Waldman and Jeffrey A. Rappaport
- Subjects
Colorectal cancer ,medicine.medical_treatment ,Receptors, Enterotoxin ,Context (language use) ,medicine.disease_cause ,030226 pharmacology & pharmacy ,Chemoprevention ,Article ,03 medical and health sciences ,0302 clinical medicine ,Medicine ,Animals ,Humans ,Pharmacology (medical) ,Molecular Targeted Therapy ,General Pharmacology, Toxicology and Pharmaceutics ,Neoplasm Staging ,business.industry ,Guanylyl Cyclase C Agonists ,General Medicine ,Immunotherapy ,Guanylate cyclase 2C ,medicine.disease ,Chimeric antigen receptor ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Plecanatide ,business ,Carcinogenesis ,Colorectal Neoplasms ,Signal Transduction - Abstract
INTRODUCTION: Colorectal cancer remains the second leading cause of cancer death in the United States, underscoring the need for novel therapies. Despite the successes of new targeted agents for other cancers, colorectal cancer suffers from a relative scarcity of actionable biomarkers. In this context, the intestinal receptor, guanylyl cyclase C (GUCY2C), has emerged as a promising target. AREAS COVERED: GUCY2C regulates a tumor suppressive signaling axis that is silenced through loss of its endogenous ligands at the earliest stages of tumorigenesis. A body of literature supports a cancer chemoprevention strategy involving reactivation of GUCY2C through FDA-approved cGMP-elevating agents such as linaclotide, plecanatide, and sildenafil. Its limited expression in extra-intestinal tissues, and retention on the surface of cancer cells, also positions GUCY2C as a target for immunotherapies to treat metastatic disease, including vaccines, chimeric antigen receptor T-cells, and antibody-drug conjugates. Likewise, GUCY2C mRNA identifies metastatic cells, enhancing colorectal cancer detection and staging. Pre-clinical and clinical programs exploring these GUCY2C-targeting strategies will be reviewed. EXPERT OPINION: Recent mechanistic insights characterizing GUCY2C ligand loss early in tumorigenesis, coupled with results from the first clinical trials testing GUCY2C-targeting strategies, continue to elevate GUCY2C as an ideal target for prevention, detection, and therapy.
- Published
- 2020
18. Cardiopoietic stem cell therapy in ischaemic heart failure: long-term clinical outcomes
- Author
-
Gerasimos Filippatos, Marco Metra, Warren Sherman, Chart Program, John R. Teerlink, William Wijns, Michal Tendera, Guy R. Heyndrickx, Roger J. Hajjar, Andre Terzic, Ricardo Sanz-Ruiz, Jozef Bartunek, Stefanie Senger, Gad Cotter, Atta Behfar, Wojciech Wojakowski, Scott A. Waldman, Thomas J. Povsic, Timothy D. Henry, Beth A. Davison, and Bernard J. Gersh
- Subjects
medicine.medical_specialty ,Cardiopoiesis ,Clinical trial ,Heart failure ,Longitudinal ,Regenerative medicine ,Stem cell ,medicine.medical_treatment ,030204 cardiovascular system & hematology ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Original Research Articles ,medicine ,Diseases of the circulatory (Cardiovascular) system ,030212 general & internal medicine ,Dosing ,Original Research Article ,business.industry ,Hazard ratio ,Stem-cell therapy ,medicine.disease ,Confidence interval ,Catheter ,RC666-701 ,Cardiology ,Cardiology and Cardiovascular Medicine ,business - Abstract
Aims This study aims to explore long‐term clinical outcomes of cardiopoiesis‐guided stem cell therapy for ischaemic heart failure assessed in the Congestive Heart Failure Cardiopoietic Regenerative Therapy (CHART‐1) trial. Methods and results CHART‐1 is a multinational, randomized, and double‐blind trial conducted in 39 centres in heart failure patients (n = 315) on standard‐of‐care therapy. The ‘active’ group received cardiopoietic stem cells delivered intramyocardially using a retention‐enhanced catheter. The ‘control’ group underwent patient‐level sham procedure. Patients were followed up to 104 weeks. In the entire study population, results of the primary hierarchical composite outcome were maintained neutral at Week 52 [Mann–Whitney estimator 0.52, 95% confidence interval (CI) 0.45–0.59, P = 0.51]. Landmark analyses suggested late clinical benefit in patients with significant left ventricular enlargement receiving adequate dosing. Specifically, beyond 100 days of follow‐up, patients with left ventricular end‐diastolic volume of 200–370 mL treated with ≤19 injections of cardiopoietic stem cells showed reduced risk of death or cardiovascular hospitalization (hazard ratio 0.38, 95% CI 0.16–0.91, P = 0.031) and cardiovascular death or heart failure hospitalization (hazard ratio 0.28, 95% CI 0.09–0.94, P = 0.040). Cardiopoietic stem cell therapy was well tolerated long term with no difference in safety readouts compared with sham at 2 years. Conclusions Longitudinal follow‐up documents that cardiopoietic stem cell therapy is overall safe, and post hoc analyses suggest benefit in an ischaemic heart failure subpopulation defined by advanced left ventricular enlargement on tolerable stem cell dosing. The long‐term clinical follow‐up thus offers guidance for future targeted trials.
- Published
- 2020
19. Chimeric Ad5.F35 vector evades anti-adenovirus serotype 5 neutralization opposing GUCY2C-targeted antitumor immunity
- Author
-
John C. Flickinger, Tingting Zhan, Trevor R. Baybutt, Jagmohan Singh, Adam E. Snook, Elinor Leong, Amanda M. Pattison, Robert D Carlson, Joshua R Barton, Ellen M. Caparosa, Babar Bashir, Scott A. Waldman, Jamin Roh, and Jeffrey A. Rappaport
- Subjects
0301 basic medicine ,Male ,Cancer Research ,medicine.medical_treatment ,viruses ,Immunology ,Receptors, Enterotoxin ,active ,immunogenicity ,immunization ,Chimerism ,Viral vector ,gastrointestinal neoplasms ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Antigen ,Immunity ,vaccine ,medicine ,Immunology and Allergy ,Animals ,Humans ,Vector (molecular biology) ,Pharmacology ,Clinical/Translational Cancer Immunotherapy ,business.industry ,Immunogenicity ,Immunotherapy ,biochemical phenomena, metabolism, and nutrition ,vaccination ,Virology ,Vaccination ,Disease Models, Animal ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Molecular Medicine ,Female ,immunotherapy ,business - Abstract
BackgroundAdenovirus serotype 5 (Ad5) is a commonly used viral vector for transient delivery of transgenes, primarily for vaccination against pathogen and tumor antigens. However, endemic infections with Ad5 produce virus-specific neutralizing antibodies (NAbs) that limit transgene delivery and constrain target-directed immunity following exposure to Ad5-based vaccines. Indeed, clinical trials have revealed the limitations that virus-specific NAbs impose on the efficacy of Ad5-based vaccines. In that context, the emerging focus on immunological approaches targeting cancer self-antigens or neoepitopes underscores the unmet therapeutic need for more efficacious vaccine vectors.MethodsHere, we evaluated the ability of a chimeric adenoviral vector (Ad5.F35) derived from the capsid of Ad5 and fiber of the rare adenovirus serotype 35 (Ad35) to induce immune responses to the tumor-associated antigen guanylyl cyclase C (GUCY2C).ResultsIn the absence of pre-existing immunity to Ad5, GUCY2C-specific T-cell responses and antitumor efficacy induced by Ad5.F35 were comparable to Ad5 in a mouse model of metastatic colorectal cancer. Furthermore, like Ad5, Ad5.F35 vector expressing GUCY2C was safe and produced no toxicity in tissues with, or without, GUCY2C expression. Importantly, this chimeric vector resisted neutralization in Ad5-immunized mice and by sera collected from patients with colorectal cancer naturally exposed to Ad5.ConclusionsThese data suggest that Ad5.F35-based vaccines targeting GUCY2C, or other tumor or pathogen antigens, may produce clinically relevant immune responses in more (≥90%) patients compared with Ad5-based vaccines (~50%).
- Published
- 2020
20. Myeloid-specific deletion of Zfp36 protects against insulin resistance and fatty liver in diet-induced obese mice
- Author
-
Caleb B. Kallen, Jason K. Kim, Robert C. Bauer, Valentina Caracciolo, Jeanette Young, Dae Young Jung, Taekyoon Kim, Hye Lim Noh, Perry J. Blackshear, Yingchun Ni, Stephen J. Flowers, Danielle O'Connell, Ross Summer, Donna M. Gonzales, and Scott A. Waldman
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Kupffer Cells ,Physiology ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Adipose tissue ,030209 endocrinology & metabolism ,Inflammation ,Diet, High-Fat ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Insulin resistance ,Tristetraprolin ,Physiology (medical) ,Diabetes mellitus ,Internal medicine ,Diabetes Mellitus ,medicine ,Animals ,Myeloid Cells ,Obesity ,RNA, Messenger ,Mice, Knockout ,Zinc finger ,business.industry ,Insulin ,Fatty liver ,Transcription Factor RelA ,Organ Size ,medicine.disease ,I-kappa B Kinase ,Fatty Liver ,030104 developmental biology ,Endocrinology ,Adipose Tissue ,Cytokines ,Insulin Resistance ,medicine.symptom ,business ,Diet-induced obese ,Research Article - Abstract
Obesity is associated with adipose tissue inflammation that contributes to insulin resistance. Zinc finger protein 36 (Zfp36) is an mRNA-binding protein that reduces inflammation by binding to cytokine transcripts and promoting their degradation. We hypothesized that myeloid-specific deficiency of Zfp36 would lead to increased adipose tissue inflammation and reduced insulin sensitivity in diet-induced obese mice. As expected, wild-type (Control) mice became obese and diabetic on a high-fat diet, and obese mice with myeloid-specific loss of Zfp36 [knockout (KO)] demonstrated increased adipose tissue and liver cytokine mRNA expression compared with Control mice. Unexpectedly, in glucose tolerance testing and hyperinsulinemic-euglycemic clamp studies, myeloid Zfp36 KO mice demonstrated improved insulin sensitivity compared with Control mice. Obese KO and Control mice had similar macrophage infiltration of the adipose depots and similar peripheral cytokine levels, but lean and obese KO mice demonstrated increased Kupffer cell (KC; the hepatic macrophage)-expressed Mac2 compared with lean Control mice. Insulin resistance in obese Control mice was associated with enhanced Zfp36 expression in KCs. Compared with Control mice, KO mice demonstrated increased hepatic mRNA expression of a multitude of classical (M1) inflammatory cytokines/chemokines, and this M1-inflammatory hepatic milieu was associated with enhanced nuclear localization of IKKβ and the p65 subunit of NF-κB. Our data confirm the important role of innate immune cells in regulating hepatic insulin sensitivity and lipid metabolism, challenge-prevailing models in which M1 inflammatory responses predict insulin resistance, and indicate that myeloid-expressed Zfp36 modulates the response to insulin in mice.
- Published
- 2018
21. Human GUCY2C-Targeted Chimeric Antigen Receptor (CAR)-Expressing T Cells Eliminate Colorectal Cancer Metastases
- Author
-
John C. Flickinger, Trevor R. Baybutt, Michael S. Magee, Priyanka Prajapati, Scott A. Waldman, Tara S. Abraham, Natalie A. Ridge, Adam R. Hersperger, Adam E. Snook, and Glen P Marszalowicz
- Subjects
Cytotoxicity, Immunologic ,0301 basic medicine ,Cancer Research ,Lung Neoplasms ,Colorectal cancer ,T-Lymphocytes ,medicine.medical_treatment ,T cell ,Immunology ,Receptors, Antigen, T-Cell ,Receptors, Enterotoxin ,Mice, Transgenic ,Immunotherapy, Adoptive ,Article ,Cell therapy ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,Cells, Cultured ,Mice, Inbred BALB C ,Receptors, Chimeric Antigen ,business.industry ,Immunotherapy ,medicine.disease ,Xenograft Model Antitumor Assays ,Chimeric antigen receptor ,HEK293 Cells ,030104 developmental biology ,Cytokine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Colorectal Neoplasms ,business - Abstract
One major hurdle to the success of adoptive T-cell therapy is the identification of antigens that permit effective targeting of tumors in the absence of toxicities to essential organs. Previous work has demonstrated that T cells engineered to express chimeric antigen receptors (CAR-T cells) targeting the murine homolog of the colorectal cancer antigen GUCY2C treat established colorectal cancer metastases, without toxicity to the normal GUCY2C-expressing intestinal epithelium, reflecting structural compartmentalization of endogenous GUCY2C to apical membranes comprising the intestinal lumen. Here, we examined the utility of a human-specific, GUCY2C-directed single-chain variable fragment as the basis for a CAR construct targeting human GUCY2C-expressing metastases. Human GUCY2C-targeted murine CAR-T cells promoted antigen-dependent T-cell activation quantified by activation marker upregulation, cytokine production, and killing of GUCY2C-expressing, but not GUCY2C-deficient, cancer cells in vitro. GUCY2C CAR-T cells provided long-term protection against lung metastases of murine colorectal cancer cells engineered to express human GUCY2C in a syngeneic mouse model. GUCY2C murine CAR-T cells recognized and killed human colorectal cancer cells endogenously expressing GUCY2C, providing durable survival in a human xenograft model in immunodeficient mice. Thus, we have identified a human GUCY2C-specific CAR-T cell therapy approach that may be developed for the treatment of GUCY2C-expressing metastatic colorectal cancer. Cancer Immunol Res; 6(5); 509–16. ©2018 AACR.
- Published
- 2018
22. Multiregion whole-exome sequencing of matched primary and metastatic tumors revealed genomic heterogeneity and suggested polyclonal seeding in colorectal cancer metastasis
- Author
-
X. Chen, Xue Zhong, Scott A. Waldman, Chun Wang, Ling Li, James Posey, J. Shu, Hushan Yang, Y. Sun, Bingshan Li, A. Yan, Zhong Ye, Atrayee Basu-Mallick, Bing-Hua Jiang, Ronald E. Myers, Lifang Hou, Danielle Fortuna, Jinliang Xing, Qiang Wei, and Juan P. Palazzo
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Colorectal cancer ,medicine.disease_cause ,Metastatic tumor ,Metastasis ,Genetic Heterogeneity ,03 medical and health sciences ,Neoplasm Seeding ,0302 clinical medicine ,Internal medicine ,Exome Sequencing ,medicine ,Humans ,Neoplasm Metastasis ,Lymph node ,Exome sequencing ,Mutation ,biology ,business.industry ,Hematology ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Polyclonal antibodies ,030220 oncology & carcinogenesis ,biology.protein ,Lymph ,Colorectal Neoplasms ,business - Abstract
Background Distant metastasis accounts for 90% of deaths from colorectal cancer (CRC). Genomic heterogeneity has been reported in various solid malignancies, but remains largely under-explored in metastatic CRC tumors, especially in primary to metastatic tumor evolution. Patients and methods We conducted high-depth whole-exome sequencing in multiple regions of matched primary and metastatic CRC tumors. Using a total of 28 tumor, normal, and lymph node tissues, we analyzed inter- and intra-individual heterogeneity, inferred the tumor subclonal architectures, and depicted the subclonal evolutionary routes from primary to metastatic tumors. Results CRC has significant inter-individual but relatively limited intra-individual heterogeneity. Genomic landscapes were more similar within primary, metastatic, or lymph node tumors than across these types. Metastatic tumors exhibited less intratumor heterogeneity than primary tumors, indicating that single-region sequencing may be adequate to identify important metastasis mutations to guide treatment. Remarkably, all metastatic tumors inherited multiple genetically distinct subclones from primary tumors, supporting a possible polyclonal seeding mechanism for metastasis. Analysis of one patient with the trio samples of primary, metastatic, and lymph node tumors supported a mechanism of synchronous parallel dissemination from the primary to metastatic tumors that was not mediated through lymph nodes. Conclusions In CRC, metastatic tumors have different but less heterogeneous genomic landscapes than primary tumors. It is possible that CRC metastasis is, at least partly, mediated through a polyclonal seeding mechanism. These findings demonstrated the rationale and feasibility for identifying and targeting primary tumor-derived metastasis-potent subclones for the prediction, prevention, and treatment of CRC metastasis.
- Published
- 2017
23. Guanylate cyclase C as a target for prevention, detection, and therapy in colorectal cancer
- Author
-
Jeffrey A. Rappaport, Adam E. Snook, Allison A. Aka, Takami Sato, Scott A. Waldman, and Amanda M. Pattison
- Subjects
0301 basic medicine ,Receptors, Peptide ,Colorectal cancer ,medicine.medical_treatment ,Receptors, Enterotoxin ,Disease ,Ligands ,medicine.disease_cause ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Biomarkers, Tumor ,medicine ,Animals ,Humans ,Gene silencing ,Pharmacology (medical) ,Molecular Targeted Therapy ,General Pharmacology, Toxicology and Pharmaceutics ,Linaclotide ,Neoplasm Staging ,business.industry ,General Medicine ,Immunotherapy ,medicine.disease ,Primary tumor ,Biomarker ,030104 developmental biology ,Receptors, Guanylate Cyclase-Coupled ,chemistry ,Drug Design ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,Colorectal Neoplasms ,Carcinogenesis ,business - Abstract
Colorectal cancer remains the second leading cause of cancer death in the United States, and new strategies to prevent, detect, and treat the disease are needed. The receptor, guanylate cyclase C (GUCY2C), a tumor suppressor expressed by the intestinal epithelium, has emerged as a promising target. Areas covered: This review outlines the role of GUCY2C in tumorigenesis, and steps to translate GUCY2C-targeting schemes to the clinic. Endogenous GUCY2C-activating ligands disappear early in tumorigenesis, silencing its signaling axis and enabling transformation. Pre-clinical models support GUCY2C ligand supplementation as a novel disease prevention paradigm. With the recent FDA approval of the GUCY2C ligand, linaclotide, and two more synthetic ligands in the pipeline, this strategy can be tested in human trials. In addition to primary tumor prevention, we also review immunotherapies targeting GUCY2C expressed by metastatic lesions, and platforms using GUCY2C as a biomarker for detection and patient staging. Expert commentary: Results of the first GUCY2C targeting schemes in patients will become available in the coming years. The identification of GUCY2C ligand loss as a requirement for colorectal tumorigenesis has the potential to change the treatment paradigm from an irreversible disease of genetic mutation, to a treatable disease of ligand insufficiency.
- Published
- 2017
24. Clinical Pharmacology & Therapeutics: Past, Present, and Future
- Author
-
Andre Terzic and Scott A. Waldman
- Subjects
Pharmacology ,Biomedical Research ,Clinical pharmacology ,Drug discovery ,business.industry ,Extramural ,030226 pharmacology & pharmacy ,Article ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,law ,030220 oncology & carcinogenesis ,Political science ,Drug Discovery ,Pharmacology, Clinical ,Health care ,Vanguard ,Humans ,Pharmacology (medical) ,Engineering ethics ,Periodicals as Topic ,business - Abstract
Clinical Pharmacology & Therapeutics (CPT), the definitive and timely source for advances in human therapeutics, transcends the drug discovery, development, regulation, and utilization continuum to catalyze, evolve, and disseminate discipline-transformative knowledge. Prioritized themes and multidisciplinary content drive the science and practice of clinical pharmacology, offering a trusted point of reference. An authoritative herald across global communities, CPT is a timeless information vehicle at the vanguard of discovery, translation, and application ushering therapeutic innovation into modern healthcare.
- Published
- 2017
25. Prenatal Regeneration in Clinical Practice
- Author
-
Scott A. Waldman
- Subjects
0301 basic medicine ,medicine.medical_specialty ,business.industry ,Regeneration (biology) ,Prenatal Care ,General Medicine ,Regenerative Medicine ,Clinical Practice ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Pregnancy ,030220 oncology & carcinogenesis ,medicine ,Humans ,Regeneration ,Female ,Hernias, Diaphragmatic, Congenital ,Intensive care medicine ,business - Published
- 2018
26. First-in-Human Use of a Retention-Enhanced Catheter for Endomyocardial Cell Delivery
- Author
-
Dariouch Dolatabadi, Jose Zefu Kimpalou, Aymeric Seron, William Wijns, Wojtek Wojakowski, Branko Beleslin, Scott A. Waldman, Andre Terzic, Guy R. Heyndrickx, Jozef Bartunek, Gerrit J. Laarman, Jean-Pierre Latere, Warren Sherman, Ricardo Sanz-Ruiz, Chart Program, and Atta Behfar
- Subjects
medicine.medical_specialty ,business.industry ,First in human ,030204 cardiovascular system & hematology ,medicine.disease ,Cell delivery ,03 medical and health sciences ,Catheter ,0302 clinical medicine ,Heart failure ,Internal medicine ,medicine ,Cardiology ,030212 general & internal medicine ,Cardiology and Cardiovascular Medicine ,business - Abstract
A novel deflectable-tip catheter (C-Cathez, Celyad, Mont-Saint-Guibert, Belgium) featuring a curved needle containing side holes developed to increase the retention of intramyocardial biotherapeutics [(1)][1] underwent first-in-human use in the CHART-1 (Congestive Heart Failure Cardiopoietic
- Published
- 2018
27. Split tolerance permits safe Ad5-GUCY2C-PADRE vaccine-induced T-cell responses in colon cancer patients
- Author
-
John C. Flickinger, Walter K. Kraft, Bo Xiang, Trevor R. Baybutt, Adam E. Snook, Scott A. Waldman, Tara S. Abraham, Takami Sato, Tingting Zhan, and Terry Hyslop
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,Male ,Cancer Research ,Receptors, Enterotoxin ,Mice ,0302 clinical medicine ,Immunogenicity, Vaccine ,Immunology and Allergy ,Cytotoxic T cell ,Vaccines, Synthetic ,biology ,Middle Aged ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Combined Modality Therapy ,3. Good health ,medicine.anatomical_structure ,GUCY2C ,Oncology ,030220 oncology & carcinogenesis ,Molecular Medicine ,Female ,Immunotherapy ,Antibody ,Colorectal Neoplasms ,Research Article ,Guanylyl cyclase C ,Colon ,T cell ,Immunology ,Genetic Vectors ,Dose-Response Relationship, Immunologic ,lcsh:RC254-282 ,Cancer Vaccines ,Adenoviridae ,03 medical and health sciences ,Immune system ,Antigen ,medicine ,Immune Tolerance ,Animals ,Humans ,Aged ,Neoplasm Staging ,Pharmacology ,business.industry ,Rectum ,Cancer ,medicine.disease ,Antibodies, Neutralizing ,Colorectal cancer ,030104 developmental biology ,biology.protein ,Cancer vaccine ,business ,Vaccine ,CD8 ,T-Lymphocytes, Cytotoxic - Abstract
Background The colorectal cancer antigen GUCY2C exhibits unique split tolerance, evoking antigen-specific CD8+, but not CD4+, T-cell responses that deliver anti-tumor immunity without autoimmunity in mice. Here, the cancer vaccine Ad5-GUCY2C-PADRE was evaluated in a first-in-man phase I clinical study of patients with early-stage colorectal cancer to assess its safety and immunological efficacy. Methods Ten patients with surgically-resected stage I or stage II (pN0) colon cancer received a single intramuscular injection of 1011 viral particles (vp) of Ad5-GUCY2C-PADRE. Safety assessment and immunomonitoring were carried out for 6 months following immunization. This trial employed continual monitoring of both efficacy and toxicity of subjects as joint primary outcomes. Results All patients receiving Ad5-GUCY2C-PADRE completed the study and none developed adverse events greater than grade 1. Antibody responses to GUCY2C were detected in 10% of patients, while 40% exhibited GUCY2C-specific T-cell responses. GUCY2C-specific responses were exclusively CD8+ cytotoxic T cells, mimicking pre-clinical studies in mice in which GUCY2C-specific CD4+ T cells are eliminated by self-tolerance, while CD8+ T cells escape tolerance and mediate antitumor immunity. Moreover, pre-existing neutralizing antibodies (NAbs) to the Ad5 vector were associated with poor vaccine-induced responses, suggesting that Ad5 NAbs oppose GUCY2C immune responses to the vaccine in patients and supported by mouse studies. Conclusions Split tolerance to GUCY2C in cancer patients can be exploited to safely generate antigen-specific cytotoxic CD8+, but not autoimmune CD4+, T cells by Ad5-GUCY2C-PADRE in the absence of pre-existing NAbs to the viral vector. Trial registration This trial (NCT01972737) was registered at ClinicalTrials.gov on October 30th, 2013. https://clinicaltrials.gov/ct2/show/NCT01972737 Electronic supplementary material The online version of this article (10.1186/s40425-019-0576-2) contains supplementary material, which is available to authorized users.
- Published
- 2018
28. GUCY2C ligand replacement to prevent colorectal cancer
- Author
-
Scott A. Waldman, Amanda M. Pattison, and Erik S. Blomain
- Subjects
0301 basic medicine ,Cancer Research ,Receptors, Peptide ,Adenoma ,Colorectal cancer ,Guanylin ,Receptors, Enterotoxin ,Context (language use) ,Review ,Ligands ,03 medical and health sciences ,chemistry.chemical_compound ,medicine ,Humans ,Receptor ,Linaclotide ,Pharmacology ,business.industry ,Guanylate cyclase 2C ,medicine.disease ,030104 developmental biology ,Receptors, Guanylate Cyclase-Coupled ,Oncology ,chemistry ,Immunology ,Cancer research ,Molecular Medicine ,Adenocarcinoma ,Colorectal Neoplasms ,business - Abstract
Despite advances in screening and prevention strategies, colorectal cancer (CRC) remains the second-leading cause of cancer-related death in the United States. Given this continued public health burden of CRC, there is a clear need for improved disease prevention. CRC initiates and progresses over decades, canonically proceeding via a series of stepwise molecular events that turn a normal epithelium into a dysfunctional epithelium, then subsequently into an adenoma, and finally an invasive adenocarcinoma. An emerging paradigm suggests that guanylyl cyclase C (GUCY2C) functions as a tumor suppressor in the intestine, and that the loss of hormone ligands for this receptor causes epithelial dysfunction and represents an important step in the disease process. In that context, GUCY2C ligand replacement therapy has been proposed as a strategy to prevent colorectal cancer, a translational opportunity that is underscored by the recent regulatory approval of the oral GUCY2C ligand linaclotide (Linzess™, Forest Laboratories and Ironwood Pharmaceuticals, Inc.).
- Published
- 2016
29. Contents Vol. 6, 2016
- Author
-
Aso Saeed, Scott A. Waldman, Yansong Zheng, Matthew S. Edwards, Timothy M. Morgan, Balakuntalam S. Kasinath, Ola Hammarsten, Tatsuru Matsukiyo, Farook Thameem, Zhilai Chen, Carlo B. Ramirez, Costas D. Lallas, Talat Tavlı, William G Hundley, Emman Shubbar, İlker Gül, Craig A. Hamilton, Ki-Bae Seung, Kiyuk Chang, Maria P. Martinez Cantarin, Pavlos Kashioulis, Ken Sakai, Ahmet Taştan, Reibin Tai, Satz Mengensatzproduktion, Gregor Guron, Yasushi Ohashi, Yury B. Lishmanov, Michael E. Hall, Pum-Joon Kim, Sobha Puppala, Scott W. Keith, Mustafa Zungur, Zhao Lin, Atsushi Aikawa, Cataldo Doria, Ravindranath Duggirala, Hun-Jun Park, Ertan Damar, Michael V. Rocco, Jennifer H. Jordan, Qiang Zeng, Ekaterina A. Alexandrova, Hanna E. Abboud, Tae Hoon Kim, Adam M. Frank, Chan Joon Kim, John Blangero, Ashesh P. Shah, Akinobu Saito, Keisuke Yamazaki, Wook Sung Chung, Bonita Falkner, Ik Jun Choi, Guilan Zhang, Druckerei Stückle, Niels Marcussen, Sang Hong Baek, Vidya S. Farook, Warren R. Maley, Zhaneta V. Vesnina, E. A. Nesterov, and John E. Hall
- Subjects
Traditional medicine ,business.industry ,Urology ,Medicine ,Cardiology and Cardiovascular Medicine ,business - Published
- 2016
30. CD19-Targeted Nanodelivery of Doxorubicin Enhances Therapeutic Efficacy in B-Cell Acute Lymphoblastic Leukemia
- Author
-
Robert W. Mason, Xinqiao Jia, Vinu Krishnan, Adam E. Snook, Dakota J Kelly, Xian Xu, Ayyappan K. Rajasekaran, and Scott A. Waldman
- Subjects
Cell Survival ,Antigens, CD19 ,Pharmaceutical Science ,Endocytosis ,Article ,CD19 ,Mice ,Drug Delivery Systems ,Cell Line, Tumor ,hemic and lymphatic diseases ,Drug Discovery ,Animals ,Humans ,Medicine ,Doxorubicin ,Cytotoxicity ,Mice, Inbred BALB C ,Acute leukemia ,biology ,business.industry ,technology, industry, and agriculture ,hemic and immune systems ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Controlled release ,Clinical trial ,Immunology ,biology.protein ,Cancer research ,Molecular Medicine ,Nanomedicine ,business ,medicine.drug - Abstract
Nanomedicine has advanced to clinical trials for adult cancer therapy. However, the field is still in its infancy for treatment of childhood malignancies such as acute lymphoblastic leukemia (ALL). Nanotherapy offers multiple advantages over conventional therapy. It facilitates targeted delivery and enables controlled release of drugs to reduce treatment-related side effects. Here, we demonstrate that doxorubicin (DOX) encapsulated in polymeric nanoparticles (NPs) modified with targeting ligands against CD19 (CD19-DOX-NPs) can be delivered in a CD19-specific manner to leukemic cells. The CD19-DOX-NPs were internalized via receptor-mediated endocytosis and imparted cytotoxicity in a CD19-dependent manner in CD19-positive ALL cells. Leukemic mice treated with CD19-DOX-NPs survived significantly longer and manifested a higher degree of agility, indicating reduced apparent systemic toxicity during treatment compared to mice treated with free DOX. We suggest that targeted delivery of drugs used in childhood cancer treatment should improve therapeutic efficacy and reduce treatment-related side effects in children.
- Published
- 2015
31. Plp1 gene duplication inhibits airway responsiveness and induces lung inflammation
- Author
-
Scott A. Waldman, Milena Hirata Armani, Yan Zhu, Thomas H. Shaffer, Lauren Sakowski, Grace M. Hobson, Portia A. Kreiger, and Elena Rodriguez
- Subjects
Male ,Pulmonary and Respiratory Medicine ,Pelizaeus-Merzbacher Disease ,Inflammation ,Respiratory physiology ,Article ,Mice ,Airway resistance ,Gene Duplication ,medicine ,Animals ,Albuterol ,Pharmacology (medical) ,Myelin Proteolipid Protein ,Methacholine Chloride ,Lung ,Dose-Response Relationship, Drug ,Interleukin-6 ,business.industry ,Biochemistry (medical) ,Interleukin ,Pelizaeus–Merzbacher disease ,Pneumonia ,medicine.disease ,Disease Models, Animal ,medicine.anatomical_structure ,Immunology ,Respiratory Mechanics ,Female ,Methacholine ,medicine.symptom ,Airway ,business ,medicine.drug - Abstract
Mice with Plp1 gene duplication model the most common form of Pelizaeus-Merzbacher disease (PMD), a CNS disease in which patients may suffer respiratory complications. We hypothesized that affected mice would lack airway responsiveness compared to wild-type and carrier mice during methacholine challenge. Wild-type (n = 10), carrier female (n = 6) and affected male (n = 8) mice were anesthetized-paralyzed, tracheostomized and ventilated. Respiratory mechanics were recorded at baseline and during escalating doses of nebulized methacholine followed by albuterol. Lung resistance (RL) was the primary endpoint. Lung tissues were assayed for inflammatory and histological differences. At baseline, phase angles were higher in carrier and affected mice than wild-type. Dose-response RL curves in affected and carrier mice indicated a lack of methacholine response. Albuterol reduced RL in wild-type and carrier, but not affected mice. Affected mice exhibited lower interleukin (IL)-6 tissue levels and alveolar inflammatory infiltrates. Affected and carrier mice, compared to wild-type, lacked airway reactivity during methacholine challenge, but only affected mice exhibited decreased lung tissue levels of IL-6 and inflammation.
- Published
- 2015
32. Editorial (Thematic Issue: Ferid Murad, at 80: A Legacy of Science, Medicine, and Mentorship)
- Author
-
Scott A. Waldman and Atta-ur-Rahman
- Subjects
Pharmacology ,Medical education ,business.industry ,Organic Chemistry ,MEDLINE ,Nitric Oxide ,Nitric oxide metabolism ,Biochemistry ,Article ,Nobel Prize ,Mentorship ,Guanylate Cyclase ,Drug Discovery ,Humans ,Molecular Medicine ,Medicine ,business ,Cyclic GMP ,Signal Transduction - Published
- 2016
33. Does obesity promote the development of colorectal cancer?
- Author
-
Scott A. Waldman and Erik S. Blomain
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Colorectal cancer ,Guanylin ,Receptors, Enterotoxin ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Risk Factors ,Internal medicine ,Animals ,Humans ,Medicine ,Pharmacology (medical) ,Obesity ,Linaclotide ,business.industry ,Guanylate cyclase 2C ,medicine.disease ,030104 developmental biology ,Endocrinology ,Oncology ,chemistry ,Cancer research ,Unfolded protein response ,Colorectal Neoplasms ,business - Published
- 2016
34. Abstracts from the 8th International Conference on cGMP Generators, Effectors and Therapeutic Implications
- Author
-
Fritz G. Rathjen, Timothy D. Calamaras, Hong Wang, Motofumi Kumazoe, Sarah Helena Nies, Jerid W. Robinson, Andreas Friebe, Balázs Tamás Németh, Michel Negrerie, Kjetil W. Andressen, Clara D. Ledsky, Sivakanan Loganathan, Kerstin Jurk, Barbara Voussen, Julia Gorelik, Mi Kyung Chang, Johannes-Peter Stasch, Friederike Cuello, Nicholas C. Blixt, S. A. Dames, Nazareno Paolocci, Gzona Bajraktari, Katherine C. Hall, Markus Waldeck-Weiermair, Marcel A. Krüger, Antonius C.F. Gorren, Donté A. Stevens, Daniel Bloch, Deborah M. Dickey, Maria Hernandez-Valladares, Lan Zhao, Alex Vincent, Jenna Scotcher, Andrzej Weichsel, Tamás Radovits, Jeremy Richman, Sophie Schobesberger, Stavros Topouzis, Regine Mühlfriedel, Dennis Busse, Reiner Frey, Sean P. Parsons, René P. Zahedi, Evanthia Mergia, Mathias W. Seeliger, Angela McLaughlin, Sylvia Nikkho, Fernando Ferreira Costa, Pavel Jansa, Ornella Manfra, Wilson A. Ferreira, Inmaculada Silos-Santiago, B. Selin Kenet, Marc Humbert, Garyfalia Makrynitsa, Katja Stehfest, Lise Román Moltzau, Shiliang Li, Chris Sarko, Lincoln R. Potter, Yusu Gu, Scott A. Waldman, Nicole Eichert, David Kilpatrick, Roland Malli, Jun Hino, Robert Solinga, Banumathi Sankaran, William R. Montfort, Alexander Kollau, Julia Davydova, Andriana Stamopoulou, Olena Rudyk, Johanna Weiss, Kenji Kangawa, Arno Fritsch, Meinrad Gawaz, Peter Sandner, Robert M. Blanton, Friedrich Grimminger, Laurinda Jaffe, Atsuko Okamoto, Elke Butt, Stefanie Peters, Christine E. Gee, Ronald J. Holewinski, Zhi Cheng Jing, Andrea Gerling, Mika Takai, Alexander Froese, Hiroshi Hosoda, Mara Goetz, Yasutake Tanaka, Ioannis I. Alexandropoulos, Amie J Moyes, Hyazinth Dobrowinski, Michael J. Shipston, Ulrike Scheib, Stefan W. Hell, Oleksandra Prysyazhna, Lukas Rüttiger, Dieter Groneberg, Vu Thao-Vi Dao, Ulrich Walter, Hariharan Subramanian, Roger Flores-Costa, Richard C. Page, Joerg Reinders, Huaqun Zhang, Dianna M. Milewicz, Martin Thunemann, Albert T. Profy, Chieri Takeuchi, Andrew B. Tobin, Darren E. Casteel, Sarah Jacobson, Philip Eaton, Cor de Witt, Nadine Mauro, Mark Aronovitz, Sevil Korkmaz-Icöz, Barbara Wilhelm, Angelos Vachaviolos, Kim Tang, Axel Gödecke, Christian Meier, Carine M. Boustany, Daniel P. Zimmer, Bernd J. Pichler, Adrian J. Hobbs, Marianne Bjørnerem, Wolfgang F. Graier, Selene J. Sollie, Jenny Tobin, Jürgen Burhenne, Meinoshin Okumura, Nancy D. Dalton, Matthias Karck, Ernst-Martin Füchtbauer, Karoline Dröbner, Kristen M. Kokkonen, Flavia Menezes, Heribert Schunkert, Kirk L. Peterson, Ying-Ju Sung, Georgios A. Spyroulias, Olga N. Petrova, Frank Eitner, Mark G. Currie, Choel Kim, Ulrike Zimmermann, Steven S. Pullen, Shunhui Zhuang, Camila B. Almeida, Sylvie Bernier, M. Wittwer, Steffen Wolter, Stylianos Michalakis, Ralf Kühn, Sara S. O. Saad, Catherine Mansfield, Joon Jung, Jennifer E. Van Eyk, Navin K. Kapur, Renate B. Pilz, David Langleben, Diane Wong, Sylvie G. Bernier, Leia C. Shuhaibar, Heinz G. Körschen, César Ibarra-Alvarado, Courtney Shea, Ryan M. Fryer, Corina Russwurm, Gilbert W. Kim, Gerald Wölkart, Marius M. Hoeper, Astrid Weiss, Harald F. Langer, Ibrahim J. Domian, Lars I. Leichert, Bernd Wissinger, Paul Allan Renhowe, Richard H. Karas, Masayuki Sasaki, Finn Olav Levy, Cor de Wit, Sonia Donzelli, Michael Russwurm, Eberhart Zrenner, Reshma S. Baliga, Holly Clifford, Jonathan C. Schisler, Cheng-Yu Chen, Nicola Conran, Jens Schlossmann, Vikas Kapil, Pablo Colorado, Boris Tchernychev, Isabelle Lamarre, Katrin Reimann, Francheska Colon-Gonzalez, Ralph T. Schermuly, Hans-Ulrich Häring, Vineet Agrawal, Peter Germano, Bernd Mayer, Katharina Beck, Moritz Lehners, Sara Vandenwijngaert, Fabíola Z. Mónica, Pavel I. Nedvetsky, Kimberly Kafadar Long, Silja Meier, Paul A. Corris, Takeshi Tokudome, Shatanik Mukherjee, Joseph R. Burgoyne, Martin Deile, Jan Giesen, Ferid Murad, G. Todd Milne, Shinpei Kawaoka, Koichi Miura, Jessica A. Wales, David A. Kass, Doris Koesling, Hongxing Chen, Frode S. Berven, Jeong Joo Kim, Friedrich W. Herberg, Kristin Hartmann, Martin Biel, Raymond L. Benza, Monte S. Willis, Daniela Bertinetti, Jaime L. Masferrer, Stefan Z. Lutz, Guang-Rong Wang, Peter Ruth, Viacheslav O. Nikolaev, Tobias Peters, Karl Ulrich Bartz-Schmidt, Arnab Ghosh, Lai Wen, Mai Kadomatsu, Sandy M. Chu, Yutaka Suzuki, Marcus Olbrich, Yigao Huang, Tobias Bauch, Thorsten Kessler, Amrita Ahluwalia, Robert Feil, Daniel Richards, Hu Sheng Qian, M Currie, Christopher Newton-Cheh, V. Kaila, Liying Qin, Peter Hegemann, Peter Wright, G-Yoon Jamie Im, Hirofumi Tachibana, Kjestine Schmidt, Pamela L. Brito, Alisa Kamynina, James C. Campbell, Adam E. Snook, Fabian Schwiering, Martin Floor, Jordi Villà-Freixa, Jana Wobst, Alexandre Dumoulin, Ali R. Banijamali, Csaba Mátyás, Harald H.H.W. Schmidt, Susanne Kohl, Mark J. Ranek, Manuela Zaccolo, Robert Lukowski, Robert Naeije, Peter P. Rainer, Stephan Rosenkranz, Emmanuel S. Buys, Simone Romoli, Andreas Papapetropoulos, Kim C. Mansky, James Wakefield, Kálmán Benke, Takashi Nojiri, Aikaterini I. Argyriou, Dongjian Hu, Glenn A. Reinhart, Cornelia Virus, Marius Ueffing, Marlies Knipper, Gerhard Hannig, Kanako Takamatsu, Athanassios Giannis, Miki Arai, Stepan Gambaryan, James E. Sheppeck, Mathew M. Mannion, S. Bruce King, Henriette Andresen, Min Zhang, Jan D. Huizinga, Jeanette Erdmann, Matthias Broser, Astrid Schrammel, Dante J. Merlino, Hossein Ardeschir Ghofrani, Gaia Calamera, Obiajulu Agha, Kathleen A Lincoln, Gerburg K. Schwaerzer, Gordon L. Warren, Andrea Neubauer, Claire Poulet, Nicoletta C. Surdo, Attila Oláh, Damian Brockschnieder, Emrah Eroglu, Carla Fernanda Franco Penteado, Walter E. Haefeli, Mihály Ruppert, Ekkehard Grünig, Konstantina Stathopoulou, Elena Walter, Gérald Simonneau, Béla Merkely, Andreas Güldner, J Keppler, Katharina Frank, Renee Sarno, Sandra Frankenreiter, Sian E. Harding, Dorit Möhrle, Paul C. Harrison, Péter Hegedűs, Hyun-Soon Geisler, James R. Klinger, Markus Wolters, Philipp Henning, Jieru E. Lin, Erik S. Blomain, Alessandro Cataliotti, Ditta Zobor, Mikiya Miyazato, Marissa Opelt, Christian Schön, John Fassett, Yue Dai, Q. Luo, Thomas G. Oertner, Linda Breci, Robert A Baumgartner, Shun Hiroi, Ping Zhang, Albert Smolenski, Paul S. Frenette, Mahmood M. Alam, Jeremy R. Egbert, Dennis J. Stuehr, Eugen Franz, Hanan Chweih, Dong I. Lee, Nadja I. Bork, Guang Liu, Hannes Schmidt, Ka Bian, Annemarie Aue, Christian Dees, Edward Seto, Manuela Harloff, Michael Paolillo Hannes Schmidt, Susanne Feil, Susanne Krasemann, Andrea Hembre Ulsund, Marina Bantzi, Laurinda A. Jaffe, Luis Agulló, Dominik M. Fischer, Wendy Baur, Kjetil Wessel Andressen, Saurav Misra, Gábor Szabó, and Oliver Pagel
- Subjects
0301 basic medicine ,Pharmacology ,03 medical and health sciences ,030104 developmental biology ,business.industry ,Pharmacology toxicology ,MEDLINE ,Medicine ,Pharmacology (medical) ,business - Published
- 2017
35. Peer Review Certifies Quality and Innovation in Clinical PharmacologyTherapeutics
- Author
-
Andre Terzic and Scott A. Waldman
- Subjects
medicine.medical_specialty ,media_common.quotation_subject ,Alternative medicine ,MEDLINE ,Bioinformatics ,030226 pharmacology & pharmacy ,Article ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,law ,medicine ,Humans ,Pharmacology (medical) ,Quality (business) ,media_common ,Pharmacology ,Publishing ,Clinical pharmacology ,business.industry ,030220 oncology & carcinogenesis ,Pharmacology, Clinical ,Engineering ethics ,Periodicals as Topic ,business ,Quality assurance - Abstract
Clinical Pharmacology & Therapeutics (CPT) is an established voice of the discipline, a trusted source of new knowledge showcasing discovery, translation and application of novel therapeutic paradigms to advance the management of patients and populations. Identifying, evaluating, prioritizing and disseminating the best science along the discovery-development-regulatory-utilization continuum are responsibilities shared through peer review. To enhance the uniformity of this essential component of quality assurance and innovation, and maximize the value of the journal and its contents to authors, reviewers, and the readership, we review key concepts concerning peer review as it specifically relates to CPT. Even if individual researchers are prone to falling in love with their own theories, the broader process of peer review and institutionalized skepticism are designed to ensure that, eventually, the best ideas prevail.Chris Mooney
- Published
- 2017
36. GUCY2C lysosomotropic endocytosis delivers immunotoxin therapy to metastatic colorectal cancer
- Author
-
Glen P Marszalowicz, Lisa D. Berman-Booty, Michael S. Magee, Adam E. Snook, Dante J. Merlino, and Scott A. Waldman
- Subjects
Colorectal cancer ,medicine.medical_treatment ,Receptors, Enterotoxin ,therapeutic targeting ,Pharmacology ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,Immunotoxin ,Medicine ,Cytotoxic T cell ,Molecular Targeted Therapy ,RNA, Small Interfering ,Gastrointestinal Neoplasms ,Drug Carriers ,0303 health sciences ,biology ,Immunotoxins ,metastatic colorectal cancer ,Antibodies, Monoclonal ,Guanylate cyclase 2C ,Endocytosis ,immunotoxin ,3. Good health ,Protein Transport ,GUCY2C ,Ricin ,Oncology ,030220 oncology & carcinogenesis ,RNA Interference ,Immunotherapy ,Antibody ,Research Paper ,Receptors, Peptide ,Caveolins ,03 medical and health sciences ,Cell Line, Tumor ,Animals ,030304 developmental biology ,business.industry ,medicine.disease ,Clathrin ,Receptors, Guanylate Cyclase-Coupled ,chemistry ,biology.protein ,Lysosomes ,business - Abstract
The emergence of targeted cancer therapy has been limited by the paucity of determinants which are tumor-specific and generally associated with disease, and have cell dynamics which effectively deploy cytotoxic payloads. Guanylyl cyclase C (GUCY2C) may be ideal for targeting because it is normally expressed only in insulated barrier compartments, including intestine and brain, but over-expressed by systemic metastatic colorectal tumors. Here, we reveal that GUCY2C rapidly internalizes from the cell surface to lysosomes in intestinal and colorectal cancer cells. Endocytosis is independent of ligand binding and receptor activation, and is mediated by clathrin. This mechanism suggests a design for immunotoxins comprising a GUCY2C-directed monoclonal antibody conjugated through a reducible disulfide linkage to ricin A chain, which is activated to a potent cytotoxin in lysosomes. Indeed, this immunotoxin specifically killed GUCY2C-expressing colorectal cancer cells in a lysosomal- and clathrin-dependent fashion. Moreover, this immunotoxin reduced pulmonary tumors >80% (p
- Published
- 2014
37. Adiponectin receptor and adiponectin signaling in human tissue among patients with end-stage renal disease
- Author
-
Maria P. Martinez Cantarin, Scott W. Keith, Scott A. Waldman, and Bonita Falkner
- Subjects
Adult ,Male ,medicine.medical_specialty ,Renal function ,Adipose tissue ,Intra-Abdominal Fat ,Real-Time Polymerase Chain Reaction ,urologic and male genital diseases ,Systemic inflammation ,CLINICAL SCIENCE ,End stage renal disease ,Insulin resistance ,Internal medicine ,medicine ,Humans ,Phosphorylation ,Cells, Cultured ,Retrospective Studies ,Adiponectin receptor 1 ,Transplantation ,Adiponectin ,business.industry ,Middle Aged ,medicine.disease ,Immunohistochemistry ,female genital diseases and pregnancy complications ,Up-Regulation ,Endocrinology ,Nephrology ,Kidney Failure, Chronic ,RNA ,Female ,Receptors, Adiponectin ,medicine.symptom ,business ,Acetyl-CoA Carboxylase ,Follow-Up Studies ,Signal Transduction ,Kidney disease - Abstract
Adiponectin plasma levels in chronic kidney disease (CKD) are two to three times higher than in individuals with normal kidney function. Despite adiponectin's anti-diabetic, anti-inflammatory and anti-atherogenic properties, patients with CKD have insulin resistance, systemic inflammation and accelerated atherogenesis. Hence, although adiponectin production is increased by adipose tissue in end-stage renal disease (ESRD), it is unclear if its effects on metabolism remain intact.To determine if there is adiponectin resistance in ESRD, we measured tissue levels of adiponectin receptor-1 (AdipoR1) and adiponectin downstream effectors in ESRD patients compared with normal kidney function controls. Blood and tissue samples were obtained from participants at the time of kidney transplantation or kidney donation. A follow-up blood sample was obtained 3-6 months after transplantation.AdipoR1 was higher in muscle and peripheral blood mononuclear cells collected from ESRD patients. There was also a nonsignificant increase in AdipoR1 in visceral fat of ESRD compared with controls. Compared with controls, phosphorylation of the adiponectin downstream effector adenosine monophosphate-activated protein kinase (AMPK) was higher in ESRD while acetyl-CoA carboxylase phosphorylation (ACC-P) and carnitine palmitoyl transferase-1 (CPT-1) levels were lower. In vitro, exposure of C2C12 cells to uremic serum resulted in upregulation of AdipoR1 and increased phosphorylation of AMPK but decreased ACC-P and CPT-1 expression.Both our in vivo and in vitro observations indicate that uremia results in upregulation of AdipoR1 but adiponectin resistance at the post-receptor level.
- Published
- 2014
38. Gut-Brain Endocrine Axes in Weight Regulation and Obesity Pharmacotherapy
- Author
-
Amanda Aing, Erik S. Blomain, Dante J. Merlino, and Scott A. Waldman
- Subjects
obesity ,business.industry ,media_common.quotation_subject ,gut-brain endocrine axis ,lcsh:R ,satiety ,lcsh:Medicine ,Appetite ,General Medicine ,Review ,Bioinformatics ,medicine.disease ,Obesity ,3. Good health ,pharmacotherapy ,Pharmacotherapy ,Endocrine system ,Medicine ,Clinical significance ,hypothalamus ,business ,media_common - Abstract
In recent years, the obesity epidemic has developed into a major health crisis both in the United States as well as throughout the developed world. With current treatments limited to expensive, high-risk surgery and minimally efficacious pharmacotherapy, new therapeutic options are urgently needed to combat this alarming trend. This review focuses on the endogenous gut-brain signaling axes that regulate appetite under physiological conditions, and discusses their clinical relevance by summarizing the clinical and preclinical studies that have investigated manipulation of these pathways to treat obesity.
- Published
- 2014
39. Managing the Innovation Supply Chain to Maximize Personalized Medicine
- Author
-
Andre Terzic and Scott A. Waldman
- Subjects
Pharmacology ,Supply chain management ,Modalities ,business.industry ,Process (engineering) ,Health Policy ,Supply chain ,Context (language use) ,Bioinformatics ,Translational Research, Biomedical ,Inventions ,Risk analysis (engineering) ,Drug Discovery ,Health care ,Humans ,DECIPHER ,Medicine ,Pharmacology (medical) ,Personalized medicine ,Diffusion of Innovation ,Precision Medicine ,business - Abstract
Personalized medicine epitomizes an evolving model of care tailored to the individual patient. This emerging paradigm harnesses radical technological advances to define each patient's molecular characteristics and decipher his or her unique pathophysiological processes. Translated into individualized algorithms, personalized medicine aims to predict, prevent, and cure disease without producing therapeutic adverse events. Although the transformative power of personalized medicine is generally recognized by physicians, patients, and payers, the complexity of translating discoveries into new modalities that transform health care is less appreciated. We often consider the flow of innovation and technology along a continuum of discovery, development, regulation, and application bridging the bench with the bedside. However, this process also can be viewed through a complementary prism, as a necessary supply chain of services and providers, each making essential contributions to the development of the final product to maximize value to consumers. Considering personalized medicine in this context of supply chain management highlights essential points of vulnerability and/or scalability that can ultimately constrain translation of the biological revolution or potentiate it into individualized diagnostics and therapeutics for optimized value creation and delivery.
- Published
- 2014
40. Blunted Evoked Prouroguanylin Endocrine Secretion in Chronic Constipation
- Author
-
Renata Tenenbaum, Patrick Griffin, Henry Foehl, Scott A. Waldman, and Peter Winkle
- Subjects
Adult ,Male ,medicine.medical_specialty ,Constipation ,media_common.quotation_subject ,Enzyme-Linked Immunosorbent Assay ,Article ,Irritable Bowel Syndrome ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Protein Precursors ,Functional GI Disorders ,Natriuretic Peptides ,Linaclotide ,Irritable bowel syndrome ,media_common ,Chronic constipation ,business.industry ,Gastroenterology ,Guanylyl Cyclase C Agonists ,Appetite ,Fasting ,Middle Aged ,medicine.disease ,3. Good health ,Endocrinology ,chemistry ,Case-Control Studies ,030220 oncology & carcinogenesis ,Plecanatide ,Female ,030211 gastroenterology & hepatology ,Nucleotides, Cyclic ,medicine.symptom ,Peptides ,business ,Receptors, Atrial Natriuretic Factor ,Uroguanylin ,Hormone - Abstract
Objectives Prouroguanylin (ProUGN) in the intestine is cleaved to form uroguanylin (UGN), which stimulates guanylate cyclase C (GUCY2C), inducing cyclic guanosine monophosphate signaling. Paracrine release regulates fluid secretion, contributing to bowel function, whereas endocrine secretion evoked by eating forms a gut-brain axis, controlling appetite. Whereas hormone insufficiency contributes to hyperphagia in obesity, its contribution to the pathophysiology of constipation syndromes remains unexplored. Here, we compared circulating ProUGN and UGN in healthy subjects and in patients with chronic idiopathic constipation (CIC) and patients with irritable bowel syndrome with constipation (IBS-C). Methods Circulating ProUGN and UGN levels were measured in 60 healthy subjects, 53 patients with CIC, and 54 patients with IBS-C. After an overnight fast, the participants ingested a standardized meal; blood samples were drawn at fasting and at 30, 60, and 90 minutes thereafter, and hormone levels were quantified by enzyme-linked immunosorbent assay. Results Fasting ProUGN levels were >30% lower in patients with CIC and those with IBS-C compared with healthy subjects regardless of age, sex, or disease state. After eating, ProUGN levels increased compared with fasting levels, although the rate of change was slower and maximum levels were lower in patients with CIC and those with IBS-C. Similarly, fasting UGN levels were lower in patients with CIC and those with IBS-C compared with healthy subjects. However, unlike ProUGN levels, UGN levels did not increase after eating. Discussion These observations support a novel pathophysiologic model in which CIC and IBS-C reflect a contribution of ProUGN insufficiency dysregulating intestinal fluid and electrolyte secretion. Translational impact This study suggests that CIC and IBS-C can be treated by oral GUCY2C hormone replacement. Indeed, these observations provide a mechanistic framework for the clinical utility of oral GUCY2C ligands like plecanatide (Trulance) and linaclotide (Linzess) to treat CIC and IBS-C.
- Published
- 2019
41. Guanylate cyclase C (GUCY2C) as a preventative and therapeutic target in colorectal cancers (CRCs) arising through divergent genomic pathways
- Author
-
Babar Bashir, Juan P. Palazzo, Scott A Waldman, Dante J. Merlino, Yeng Fing, Jeffrey A. Rappaport, Adam Snook, Eric R. Fearon, and Esteban Gnass
- Subjects
Cancer Research ,Oncology ,business.industry ,Cancer research ,Medicine ,Guanylate cyclase C ,business ,CDX2 ,neoplasms ,digestive system diseases ,Epigenetic silencing - Abstract
595 Background: CRCs arise through distinct mutations, including in APC pathway leading to tubular adenomas (TAs); in BRAF, with epigenetic silencing of CDX2, leading to serrated adenomas (SAs); and in the DNA mismatch repair machinery driving microsatellite instability (MSI). The APC pathway involves loss of the hormone guanylin, silencing the tumor suppressing receptor GUCY2C. Indeed, oral hormone replacement is an emerging strategy to reactivate GUCY2C and prevent CRC. Moreover, retained expression by tumors arising from TAs has established GUCY2C as a therapeutic target to prevent and treat metastatic CRC. Here, we defined the potential role of the guanylin-GUCY2C axis, and its suitability as a target, in tumors arising through the SA and MSI pathways. Methods: We compared guanylin-GUCY2C protein and mRNA expression between human TAs (n = 18), SAs (n = 15), MSI tumors (n = 7) and their matched normal adjacent tissue. Genetic mouse models of serrated and MSI tumors were used to confirm findings and elucidate mechanisms. Results: Guanylin hormone was eliminated in TAs, SAs and MSI tumors compared to their normal adjacent tissues. In contrast to the hormone, the tumor suppressing receptor GUCY2C was retained in TAs and MSI tumors. Surprisingly, GUCY2C expression was nearly eliminated in SAs reflecting loss of the transcription factor CDX2. Changes in the guanylin-GUCY2C axis in human SAs and MSI tumors were precisely recapitulated in genetic mouse models. Conclusions: Guanylin is universally lost at the earliest stages of transformation in tumors arising through divergent genomic mechanisms suggesting its utility as a biomarker of CRC initiation. These data reveal the possibility of guanylin loss silencing GUCY2C in the pathophysiology of, and oral hormone replacement to restore GUCY2C signaling to prevent, MSI tumors. Also, they highlight the potential for targeting GUCY2C to prevent and treat metastases arising from TAs and MSI tumors. In contrast, loss of GUCY2C excludes patients with SAs as candidates for GUCY2C-based prevention and therapy.
- Published
- 2019
42. Genetics and Genomics for the Prevention and Treatment of Cardiovascular Disease: Update
- Author
-
Santhi K, Ganesh, Donna K, Arnett, Themistocles L, Assimes, Thermistocles L, Assimes, Craig T, Basson, Aravinda, Chakravarti, Patrick T, Ellinor, Mary B, Engler, Elizabeth, Goldmuntz, David M, Herrington, Ray E, Hershberger, Yuling, Hong, Julie A, Johnson, Steven J, Kittner, Deborah A, McDermott, James F, Meschia, Luisa, Mestroni, Christopher J, O'Donnell, Bruce M, Psaty, Ramachandran S, Vasan, Marc, Ruel, Win-Kuang, Shen, Andre, Terzic, and Scott A, Waldman
- Subjects
Heart disease ,Population ,Genomics ,Disease ,Bioinformatics ,Physiology (medical) ,Genetics ,medicine ,Humans ,education ,Genetic testing ,education.field_of_study ,medicine.diagnostic_test ,business.industry ,Genetic heterogeneity ,American Heart Association ,medicine.disease ,United States ,Genetic architecture ,Phenotype ,Cardiovascular Diseases ,Pharmacogenetics ,Mutation ,Cardiology and Cardiovascular Medicine ,business - Abstract
Cardiovascular diseases (CVDs) are a major source of morbidity and mortality worldwide. Despite a decline of ≈30% over the past decade, heart disease remains the leading killer of Americans.1 For rare and familial forms of CVD, we are increasingly recognizing single-gene mutations that impart relatively large effects on individual phenotype. Examples include inherited forms of cardiomyopathy, arrhythmias, and aortic diseases. However, the prevalence of monogenic disorders typically accounts for a small proportion of the total CVD observed in the population. CVDs in the general population are complex diseases, with several contributing genetic and environmental factors. Although recent progress in monogenic disorders has occurred, we have seen a period of intense investigation to identify the genetic architecture of more common forms of CVD and related traits. Genomics serves several roles in cardiovascular health and disease, including disease prediction, discovery of genetic loci influencing CVD, functional evaluation of these genetic loci to understand mechanisms, and identification of therapeutic targets. For single-gene CVDs, progress has led to several clinically useful diagnostic tests, extending our ability to inform the management of afflicted patients and their family members. However, there has been little progress in developing genetic testing for complex CVD because individual common variants have only a modest impact on risk. The study of the genomics of complex CVDs is further challenged by the influence of environmental variables, phenotypic heterogeneity, and pathogenic complexity. Characterization of the clinical phenotype requires consideration of the clinical details of the diseases and traits under study. This update expands the prior scientific statement on the relevance of genetics and genomics for the prevention and treatment of CVDs.2 In the earlier report, we focused on the current status of the field, which consisted of predominantly family-based linkage studies and single-gene or mendelian mutations of relatively large phenotypic effect …
- Published
- 2013
43. Companion diagnostics at the intersection of personalized medicine and healthcare delivery
- Author
-
Scott A. Waldman and Andre Terzic
- Subjects
business.industry ,Proteomic Profiling ,Biochemistry (medical) ,Clinical Biochemistry ,Disease ,Evidence-based medicine ,Molecular diagnostics ,Bioinformatics ,Precision medicine ,Data science ,Article ,Drug Discovery ,Health care ,Profiling (information science) ,Medicine ,Personalized medicine ,business - Abstract
Progress in understanding biological circuits, advances in enabling technologies including the high-throughput platforms of genomics, proteomics and metabolomics, the evolution in drug target discovery and the development of companion diagnostics set the healthcare enterprise on the verge of personalized disease management [1]. This revolution in clinical care is dependent on molecular diagnostics that predict and prevent disease, enabling the diagnosis and treatment of individual patients and populations [2]. Diagnostic biomarkers are quantifiable disease characteristics that provide information about underlying molecular processes to define disease progression or predict treatment response. Familiar diagnostic biomarkers include traditional measurements (heart rate and blood pressure), imaging techniques (chest x-ray and mammograms) and protein measurements (PSA and CEA). The revolution in biology and high-throughput technology has provided an opportunity to develop a new generation of companion and complementary diagnostics, including single-nucleotide polymorphism analysis, genomic and proteomic profiling, epigenetic profiling and gene expression profiling [3]. In turn, these diagnostics increase disease-specific sensitivity and specificity, contributing to the accuracy of personalized disease management. This advancing wave of innovation has induced the next generation of biotechnology to capture the use of companion diagnostics for the application of specific therapeutic agents to the clinical care of individuals and populations [4]. Yet, as pointed out in this issue by Milne et al., the potential of biomarker technologies, in the form of companion and complementary diagnostics, to revolutionize clinical care has not been fully realized, reflecting a disconnect between the emergence of discovery technologies and models for their validation, early adoption and application across disease populations [5]. These limitations in the validation of molecular diagnostics have raised considerations regarding approval and marketing by regulatory agencies. Moreover, as highlighted in this issue by Cohen et al., the paucity of biomarker validation serves as a considerable obstacle to the adoption of companion diagnostics by healthcare providers and payors [6]. The evolving regulatory and reimbursement environments, in conjunction with the central importance of analytic validation and clinical qualification, has resulted in barriers to adoption that have restricted the full impact of companion diagnostics in clinical practice [5,6]. The emergence of analytic technologies for evaluating nucleic acids and proteins, which are associated with the deconvolution of the human genome, provided the technological push to develop molecular biomarkers for disease management [7]. Conversely, Companion diagnostics at the intersection of personalized medicine and healthcare delivery
- Published
- 2015
44. Cardiopoietic Stem Cell Therapy in Heart Failure
- Author
-
Michal Tendera, Scott A. Waldman, Dariouch Dolatabadi, Marko Banovic, C.J. Vrints, William Wijns, Badih El Nakadi, Christian Homsy, Jozef Bartunek, Mathias Vrolix, Atta Behfar, Andre Terzic, Victor Legrand, Branko Beleslin, Jean-Louis Vanoverschelde, Marc Vanderheyden, Miodrag Ostojic, Jo Dens, and Ruben J. Crespo-Diaz
- Subjects
Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,030204 cardiovascular system & hematology ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,law ,Internal medicine ,medicine ,030304 developmental biology ,0303 health sciences ,Ischemic cardiomyopathy ,Ejection fraction ,business.industry ,Stem-cell therapy ,medicine.disease ,3. Good health ,Surgery ,Clinical trial ,Heart failure ,Human medicine ,Stem cell ,business ,Cardiology and Cardiovascular Medicine - Abstract
Objectives This study sought to evaluate the feasibility and safety of autologous bone marrow-derived and cardiogenically oriented mesenchymal stem cell therapy and to probe for signs of efficacy in patients with chronic heart failure. Background In pre-clinical heart failure models, cardiopoietic stem cell therapy improves left ventricular function and blunts pathological remodeling. Methods The C-CURE (Cardiopoietic stem Cell therapy in heart failURE) trial, a prospective, multicenter, randomized trial, was conducted in patients with heart failure of ischemic origin who received standard of care or standard of care plus lineage-specified stem cells. In the cell therapy arm, bone marrow was harvested and isolated mesenchymal stem cells were exposed to a cardiogenic cocktail. Derived cardiopoietic stem cells, meeting release criteria under Good Manufacturing Practice, were delivered by endomyocardial injections guided by left ventricular electromechanical mapping. Data acquisition and analysis were performed in blinded fashion. The primary endpoint was feasibility/safety at 2-year follow-up. Secondary endpoints included cardiac structure/function and measures of global clinical performance 6 months post-therapy. Results Mesenchymal stem cell cocktail-based priming was achieved for each patient with the dose attained in 75% and delivery without complications in 100% of cases. There was no evidence of increased cardiac or systemic toxicity induced by cardiopoietic cell therapy. Left ventricular ejection fraction was improved by cell therapy (from 27.5 +/- 1.0% to 34.5 +/- 1.1%) versus standard of care alone (from 27.8 +/- 2.0% to 28.0 +/- 1.8%, p < 0.0001) and was associated with a reduction in left ventricular end-systolic volume (-24.8 +/- 3.0 ml vs. -8.8 +/- 3.9 ml, p < 0.001). Cell therapy also improved the 6-min walk distance (+62 +/- 18 m vs. -15 +/- 20 m, p < 0.01) and provided a superior composite clinical score encompassing cardiac parameters in tandem with New York Heart Association functional class, quality of life, physical performance, hospitalization, and event-free survival. Conclusions The C-CURE trial implements the paradigm of lineage guidance in cell therapy. Cardiopoietic stem cell therapy was found feasible and safe with signs of benefit in chronic heart failure, meriting definitive clinical evaluation. (C-Cure Clinical Trial; NCT00810238) (C) 2013 by the American College of Cardiology Foundation
- Published
- 2013
- Full Text
- View/download PDF
45. Molecular Staging of Node Negative Patients with Colorectal Cancer
- Author
-
Scott A. Waldman and Terry Hyslop
- Subjects
Oncology ,Guanylyl cyclase C ,medicine.medical_specialty ,Pathology ,Colorectal cancer ,Context (language use) ,Disease ,Review ,Molecular stage ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,030212 general & internal medicine ,Lymph node ,business.industry ,Early detection ,medicine.disease ,Occult ,3. Good health ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,Cancer biomarkers ,Lymph ,business - Abstract
Metastatic disease is the principle cause of death from colorectal cancer. In that context, the most significant indicator of overall survival and therapeutic response to adjuvant chemotherapy is the presence of metastatic tumor cells in regional lymph nodes. Although histopathologic analysis of lymph nodes is central to all colorectal cancer staging paradigms, its prognostic and predictive value is limited. Indeed, about 30% of patients with histopathology-negative lymph nodes (pN0) die from metastatic disease, reflected by microscopic lymph node metastases that are overlooked by standard techniques. These unrecognized tumor cells are especially important when considering racial disparities in outcomes in colorectal cancer patients, where blacks with lymph node-negative disease have the largest discrepancies in outcomes, with more than 40% excess mortality compared to Caucasian patients. However, the significance of tumor cells in regional lymph nodes remains uncertain, and approximately 50% of colorectal cancer patients with nodal metastases detected by histopathology remain free of recurrent disease. Accurate identification of occult metastases in regional lymph nodes, and defining their value as prognostic markers of recurrence risk and predictive markers of response to adjuvant chemotherapy remains one challenge in the management of colorectal cancer patients. Guanylyl cyclase C (GUCY2C), a receptor which is expressed primarily in intestinal cells normally, but is universally over-expressed by colorectal cancer cells, has been validated to detect prognostically significant occult metastases using quantitative RT-PCR (RT-qPCR). Biomarker validation was achieved through a prospective, multicenter, blinded clinical trial. In that trial, occult tumor burden estimated across all regional lymph nodes by GUCY2C RT-qPCR predicted clinical outcomes, identifying node-negative patients with a low (near zero) risk, and those with >80% risk, of developing disease recurrence. Moreover, there was disproportionately higher occult tumor burden in black, compared to white, patients which contributes to racial disparities in outcomes in colorectal cancer. The diagnostic paradigm quantifying occult tumor burden using GUCY2C qRT-PCR is positioned to reduce racial disparities in colorectal cancer mortality.
- Published
- 2013
46. Guanylyl cyclase C as a biomarker in colorectal cancer
- Author
-
Terry Hyslop and Scott A. Waldman
- Subjects
Oncology ,medicine.medical_specialty ,Pathology ,Receptors, Peptide ,Colorectal cancer ,Clinical Biochemistry ,Receptors, Enterotoxin ,Disease ,Colon cancer staging ,Article ,Internal medicine ,Drug Discovery ,Biomarkers, Tumor ,medicine ,Humans ,Disseminated disease ,Neoplasm Staging ,business.industry ,Biochemistry (medical) ,Guanylate cyclase 2C ,medicine.disease ,Occult ,Receptors, Guanylate Cyclase-Coupled ,Lymphatic Metastasis ,Biomarker (medicine) ,Lymph Nodes ,Lymph ,Colorectal Neoplasms ,business - Abstract
While histologic assessment of nodes is a component of all colon cancer staging paradigms, approximately 30% of patients with histology-negative nodes (pN0) die of disseminated disease reflected by occult nodal metastases. Undetected metastases are particularly important when considering racial disparities in colon cancer, where black subjects with pN0 disease exhibit the greatest differences in outcomes, with >40% excess mortality. Recently, guanylyl cyclase C (GCC), a protein normally restricted to intestinal cells, but universally expressed by colorectal cancer cells, was validated for detecting occult metastases. Indeed, occult tumor burden across regional lymph nodes estimated by GCC quantitative reverse transcription PCR identifies pN0 patients with near zero risk, and those with >80% risk, of unfavorable outcomes. Disproportionately high occult tumor burden in black patients underlies racial disparities in stage-specific mortality. These studies position the platform encompassing quantification of occult tumor burden by GCC quantitative reverse transcription PCR for translation, as a detect–treat paradigm to reduce racial disparities in colon cancer mortality.
- Published
- 2013
47. Obesity pharmacotherapy: What is next?
- Author
-
Gilbert W. Kim, Scott A. Waldman, Michael A. Valentino, Francheska Colon-Gonzalez, and Jieru E. Lin
- Subjects
medicine.medical_specialty ,Clinical Biochemistry ,Population ,Appetite ,Pharmacology ,Overweight ,Synaptic Transmission ,Biochemistry ,Article ,Drug Delivery Systems ,Pharmacotherapy ,Anti-Obesity Agents ,Weight loss ,Weight Loss ,Weight management ,medicine ,Humans ,Obesity ,education ,Intensive care medicine ,Life Style ,Molecular Biology ,education.field_of_study ,business.industry ,Neuropeptides ,Lipase ,General Medicine ,Pancreatic Hormones ,medicine.disease ,Drug development ,Molecular Medicine ,medicine.symptom ,business - Abstract
The increase in obesity in the Unites States and around the world in the last decade is overwhelming. The number of overweight adults in the world surpassed 1 billion in 2008. Health hazards associated with obesity are serious and include heart disease, sleep apnea, diabetes, and cancer. Although lifestyle modifications are the most straightforward way to control weight, a large portion of the population may not be able to rely on this modality alone. Thus, the development of anti-obesity therapeutics represents a major unmet medical need. Historically, anti-obesity pharmacotherapies have been unsafe and minimally efficacious. A better understanding of the biology of appetite and metabolism provides an opportunity to develop drugs that may offer safer and more effective alternatives for weight management. This review discusses drugs that are currently on the market and in development as anti-obesity therapeutics based on their target and mechanism of action. It should serve as a roadmap to establish expectations for the near future for anti-obesity drug development.
- Published
- 2013
48. Evidence-based Guidelines for Precision Risk Stratification-Based Screening (PRSBS) for Colorectal Cancer: Lessons learned from the US Armed Forces: Consensus and Future Directions
- Author
-
Patrick E. Young, Yan Gao Man, Itzhak Avital, Judy Yee, Vadim Backman, Mladjan Protic, Aviram Nissan, Khristian Noto, Martin Daumer, Warren S. Grundfest, Renee Mueller, Russell C. Langan, Thomas A. Summers, Craig Womeldorph, Scott R. Steele, Björn L.D.M. Brücher, Anton J. Bilchik, John Eberhardt, Alexander Stojadinovic, Paul Mancusco, and Scott A. Waldman
- Subjects
medicine.medical_specialty ,Evidence-based practice ,colon ,business.industry ,Colorectal cancer ,rectal ,Ethnic group ,Cancer ,colorectal cancer ,Review ,Evidence-based medicine ,Disease ,Bioinformatics ,medicine.disease ,risk identification ,Oncology ,cancer screening ,consensus ,Cancer screening ,medicine ,evidence-based medicine ,Intensive care medicine ,business ,Socioeconomic status - Abstract
Colorectal cancer (CRC) is the third most common cause of cancer-related death in the United States (U.S.), with estimates of 143,460 new cases and 51,690 deaths for the year 2012. Numerous organizations have published guidelines for CRC screening; however, these numerical estimates of incidence and disease-specific mortality have remained stable from years prior. Technological, genetic profiling, molecular and surgical advances in our modern era should allow us to improve risk stratification of patients with CRC and identify those who may benefit from preventive measures, early aggressive treatment, alternative treatment strategies, and/or frequent surveillance for the early detection of disease recurrence. To better negotiate future economic constraints and enhance patient outcomes, ultimately, we propose to apply the principals of personalized and precise cancer care to risk-stratify patients for CRC screening (Precision Risk Stratification-Based Screening, PRSBS). We believe that genetic, molecular, ethnic and socioeconomic disparities impact oncological outcomes in general, those related to CRC, in particular. This document highlights evidence-based screening recommendations and risk stratification methods in response to our CRC working group private-public consensus meeting held in March 2012. Our aim was to address how we could improve CRC risk stratification-based screening, and to provide a vision for the future to achieving superior survival rates for patients diagnosed with CRC.
- Published
- 2013
49. Bioactivity of Oral Linaclotide in Human Colorectum for Cancer Chemoprevention
- Author
-
Jieru E. Lin, Drew K. Seisler, Leo Katz, Asad Umar, Gary Della'Zanna, Paul J. Limburg, Scott A. Waldman, David Kastenberg, Nathan R. Foster, David S. Weinberg, and Walter K. Kraft
- Subjects
0301 basic medicine ,Agonist ,Cancer Research ,medicine.medical_specialty ,medicine.drug_class ,Colon ,Guanylin ,Rectum ,Administration, Oral ,Receptors, Enterotoxin ,Pharmacology ,Article ,Polyethylene Glycols ,Gastrointestinal Hormones ,03 medical and health sciences ,chemistry.chemical_compound ,Cecum ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,Humans ,Large intestine ,Phosphorylation ,Natriuretic Peptides ,Linaclotide ,Cyclic GMP ,business.industry ,Microfilament Proteins ,Cancer ,Epithelial Cells ,Guanylyl Cyclase C Agonists ,Colonoscopy ,medicine.disease ,Phosphoproteins ,Healthy Volunteers ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Ki-67 Antigen ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,business ,Colorectal Neoplasms ,Peptides ,Cell Adhesion Molecules ,Uroguanylin - Abstract
Guanylate cyclase C (GUCY2C) is a tumor-suppressing receptor silenced by loss of expression of its luminocrine hormones guanylin and uroguanylin early in colorectal carcinogenesis. This observation suggests oral replacement with a GUCY2C agonist may be an effective targeted chemoprevention agent. Linaclotide is an FDA-approved oral GUCY2C agonist formulated for gastric release, inducing fluid secretion into the small bowel to treat chronic idiopathic constipation. The ability of oral linaclotide to induce a pharmacodynamic response in epithelial cells of the colorectum in humans remains undefined. Here, we demonstrate that administration of 0.87 mg of oral linaclotide daily for 7 days to healthy volunteers, after oral colon preparation with polyethylene glycol solution (MoviPrep), activates GUCY2C, resulting in accumulation of its product cyclic (c)GMP in epithelial cells of the cecum, transverse colon, and distal rectum. GUCY2C activation by oral linaclotide was associated with homeostatic signaling, including phosphorylation of vasodilator-stimulated phosphoprotein and inhibition of proliferation quantified by reduced Ki67-positive epithelial cells. In the absence of the complete oral colonoscopy preparation, linaclotide did not alter cGMP production in epithelial cells of the colorectum, demonstrating that there was an effect related to the laxative preparation. These data show that the current FDA-approved formulation of oral linaclotide developed for small-bowel delivery to treat chronic idiopathic constipation is inadequate for reliably regulating GUCY2C in the colorectum to prevent tumorigenesis. The study results highlight the importance of developing a novel GUCY2C agonist formulated for release and activity targeted to the large intestine for colorectal cancer prevention. Cancer Prev Res; 10(6); 345–54. ©2017 AACR.
- Published
- 2016
50. 31st Annual Meeting and Associated Programs of the Society for Immunotherapy of Cancer (SITC 2016): part two
- Author
-
Casey Ager, Matthew Reilley, Courtney Nicholas, Todd Bartkowiak, Ashvin Jaiswal, Michael Curran, Tina C. Albershardt, Anshika Bajaj, Jacob F. Archer, Rebecca S. Reeves, Lisa Y. Ngo, Peter Berglund, Jan ter Meulen, Caroline Denis, Hormas Ghadially, Thomas Arnoux, Fabien Chanuc, Nicolas Fuseri, Robert W. Wilkinson, Nicolai Wagtmann, Yannis Morel, Pascale Andre, Michael B. Atkins, Matteo S. Carlino, Antoni Ribas, John A. Thompson, Toni K. Choueiri, F. Stephen Hodi, Wen-Jen Hwu, David F. McDermott, Victoria Atkinson, Jonathan S. Cebon, Bernie Fitzharris, Michael B. Jameson, Catriona McNeil, Andrew G. Hill, Eric Mangin, Malidi Ahamadi, Marianne van Vugt, Mariëlle van Zutphen, Nageatte Ibrahim, Georgina V. Long, Robyn Gartrell, Zoe Blake, Ines Simoes, Yichun Fu, Takuro Saito, Yingzhi Qian, Yan Lu, Yvonne M. Saenger, Sadna Budhu, Olivier De Henau, Roberta Zappasodi, Kyle Schlunegger, Bruce Freimark, Jeff Hutchins, Christopher A. Barker, Jedd D. Wolchok, Taha Merghoub, Elena Burova, Omaira Allbritton, Peter Hong, Jie Dai, Jerry Pei, Matt Liu, Joel Kantrowitz, Venus Lai, William Poueymirou, Douglas MacDonald, Ella Ioffe, Markus Mohrs, William Olson, Gavin Thurston, Cristian Capasso, Federica Frascaro, Sara Carpi, Siri Tähtinen, Sara Feola, Manlio Fusciello, Karita Peltonen, Beatriz Martins, Madeleine Sjöberg, Sari Pesonen, Tuuli Ranki, Lukasz Kyruk, Erkko Ylösmäki, Vincenzo Cerullo, Fabio Cerignoli, Biao Xi, Garret Guenther, Naichen Yu, Lincoln Muir, Leyna Zhao, Yama Abassi, Víctor Cervera-Carrascón, Mikko Siurala, João Santos, Riikka Havunen, Suvi Parviainen, Akseli Hemminki, Angus Dalgleish, Satvinder Mudan, Mark DeBenedette, Ana Plachco, Alicia Gamble, Elizabeth W. Grogan, John Krisko, Irina Tcherepanova, Charles Nicolette, Pooja Dhupkar, Ling Yu, Eugenie S. Kleinerman, Nancy Gordon, Italia Grenga, Lauren Lepone, Sofia Gameiro, Karin M. Knudson, Massimo Fantini, Kwong Tsang, James Hodge, Renee Donahue, Jeffrey Schlom, Elizabeth Evans, Holm Bussler, Crystal Mallow, Christine Reilly, Sebold Torno, Maria Scrivens, Cathie Foster, Alan Howell, Leslie Balch, Alyssa Knapp, John E. Leonard, Mark Paris, Terry Fisher, Siwen Hu-Lieskovan, Ernest Smith, Maurice Zauderer, William Fogler, Marilyn Franklin, Matt Thayer, Dan Saims, John L. Magnani, Jian Gong, Michael Gray, George Fromm, Suresh de Silva, Louise Giffin, Xin Xu, Jason Rose, Taylor H. Schreiber, Sofia R. Gameiro, Paul E. Clavijo, Clint T. Allen, James W. Hodge, Kwong Y. Tsang, Jane Grogan, Nicholas Manieri, Eugene Chiang, Patrick Caplazi, Mahesh Yadav, Patrick Hagner, Hsiling Chiu, Michelle Waldman, Anke Klippel, Anjan Thakurta, Michael Pourdehnad, Anita Gandhi, Ian Henrich, Laura Quick, Rob Young, Margaret Chou, Andrew Hotson, Stephen Willingham, Po Ho, Carmen Choy, Ginna Laport, Ian McCaffery, Richard Miller, Kimberly A. Tipton, Kenneth R. Wong, Victoria Singson, Chihunt Wong, Chanty Chan, Yuanhiu Huang, Shouchun Liu, Jennifer H. Richardson, W. Michael Kavanaugh, James West, Bryan A. Irving, Ritika Jaini, Matthew Loya, Charis Eng, Melissa L. Johnson, Alex A. Adjei, Mateusz Opyrchal, Suresh Ramalingam, Pasi A. Janne, George Dominguez, Dmitry Gabrilovich, Laura de Leon, Jeannette Hasapidis, Scott J. Diede, Peter Ordentlich, Scott Cruickshank, Michael L. Meyers, Matthew D. Hellmann, Pawel Kalinski, Amer Zureikat, Robert Edwards, Ravi Muthuswamy, Nataša Obermajer, Julie Urban, Lisa H. Butterfield, William Gooding, Herbert Zeh, David Bartlett, Olga Zubkova, Larissa Agapova, Marina Kapralova, Liudmila Krasovskaia, Armen Ovsepyan, Maxim Lykov, Artem Eremeev, Vladimir Bokovanov, Olga Grigoryeva, Andrey Karpov, Sergey Ruchko, Alexandr Shuster, Danny N. Khalil, Luis Felipe Campesato, Yanyun Li, Adam S. Lazorchak, Troy D. Patterson, Yueyun Ding, Pottayil Sasikumar, Naremaddepalli Sudarshan, Nagaraj Gowda, Raghuveer Ramachandra, Dodheri Samiulla, Sanjeev Giri, Rajesh Eswarappa, Murali Ramachandra, David Tuck, Timothy Wyant, Jasmin Leshem, Xiu-fen Liu, Tapan Bera, Masaki Terabe, Birgit Bossenmaier, Gerhard Niederfellner, Yoram Reiter, Ira Pastan, Leiming Xia, Yang Xia, Yangyang Hu, Yi Wang, Yangyi Bao, Fu Dai, Shiang Huang, Elaine Hurt, Robert E. Hollingsworth, Lawrence G. Lum, Alfred E. Chang, Max S. Wicha, Qiao Li, Thomas Mace, Neil Makhijani, Erin Talbert, Gregory Young, Denis Guttridge, Darwin Conwell, Gregory B. Lesinski, Rodney JM Macedo Gonzales, Austin P. Huffman, Ximi K. Wang, Ran Reshef, Andy MacKinnon, Jason Chen, Matt Gross, Gisele Marguier, Peter Shwonek, Natalija Sotirovska, Susanne Steggerda, Francesco Parlati, Amani Makkouk, Mark K. Bennett, Ethan Emberley, Tony Huang, Weiqun Li, Silinda Neou, Alison Pan, Jing Zhang, Winter Zhang, Netonia Marshall, Thomas U. Marron, Judith Agudo, Brian Brown, Joshua Brody, Christopher McQuinn, Matthew Farren, Hannah Komar, Reena Shakya, Thomas Ludwug, Y. Maurice Morillon, Scott A. Hammond, John W. Greiner, Pulak R. Nath, Anthony L. Schwartz, Dragan Maric, David D. Roberts, Aung Naing, Kyriakos P. Papadopoulos, Karen A. Autio, Deborah J. Wong, Manish Patel, Gerald Falchook, Shubham Pant, Patrick A. Ott, Melinda Whiteside, Amita Patnaik, John Mumm, Filip Janku, Ivan Chan, Todd Bauer, Rivka Colen, Peter VanVlasselaer, Gail L. Brown, Nizar M. Tannir, Martin Oft, Jeffrey Infante, Evan Lipson, Ajay Gopal, Sattva S. Neelapu, Philippe Armand, Stephen Spurgeon, John P. Leonard, Rachel E. Sanborn, Ignacio Melero, Thomas F. Gajewski, Matthew Maurer, Serena Perna, Andres A. Gutierrez, Raphael Clynes, Priyam Mitra, Satyendra Suryawanshi, Douglas Gladstone, Margaret K. Callahan, James Crooks, Sheila Brown, Audrey Gauthier, Marc Hillairet de Boisferon, Andrew MacDonald, Laura Rosa Brunet, William T. Rothwell, Peter Bell, James M. Wilson, Fumi Sato-Kaneko, Shiyin Yao, Shannon S. Zhang, Dennis A. Carson, Cristina Guiducci, Robert L. Coffman, Kazutaka Kitaura, Takaji Matsutani, Ryuji Suzuki, Tomoko Hayashi, Ezra E. W. Cohen, David Schaer, Yanxia Li, Julie Dobkin, Michael Amatulli, Gerald Hall, Thompson Doman, Jason Manro, Frank Charles Dorsey, Lillian Sams, Rikke Holmgaard, Krishnadatt Persaud, Dale Ludwig, David Surguladze, John S. Kauh, Ruslan Novosiadly, Michael Kalos, Kyla Driscoll, Hardev Pandha, Christy Ralph, Kevin Harrington, Brendan Curti, Wallace Akerley, Sumati Gupta, Alan Melcher, David Mansfield, David R. Kaufman, Emmett Schmidt, Mark Grose, Bronwyn Davies, Roberta Karpathy, Darren Shafren, Katerina Shamalov, Cyrille Cohen, Naveen Sharma, James Allison, Tala Shekarian, Sandrine Valsesia-Wittmann, Christophe Caux, Aurelien Marabelle, Brian M. Slomovitz, Kathleen M. Moore, Hagop Youssoufian, Marshall Posner, Poonam Tewary, Alan D. Brooks, Ya-Ming Xu, Kithsiri Wijeratne, Leslie A. A. Gunatilaka, Thomas J. Sayers, John P. Vasilakos, Tesha Alston, Simon Dovedi, James Elvecrog, Iwen Grigsby, Ronald Herbst, Karen Johnson, Craig Moeckly, Stefanie Mullins, Kristen Siebenaler, Julius SternJohn, Ashenafi Tilahun, Mark A. Tomai, Katharina Vogel, Eveline E. Vietsch, Anton Wellstein, Martin Wythes, Stefano Crosignani, Joseph Tumang, Shilpa Alekar, Patrick Bingham, Sandra Cauwenberghs, Jenny Chaplin, Deepak Dalvie, Sofie Denies, Coraline De Maeseneire, JunLi Feng, Kim Frederix, Samantha Greasley, Jie Guo, James Hardwick, Stephen Kaiser, Katti Jessen, Erick Kindt, Marie-Claire Letellier, Wenlin Li, Karen Maegley, Reece Marillier, Nichol Miller, Brion Murray, Romain Pirson, Julie Preillon, Virginie Rabolli, Chad Ray, Kevin Ryan, Stephanie Scales, Jay Srirangam, Jim Solowiej, Al Stewart, Nicole Streiner, Vince Torti, Konstantinos Tsaparikos, Xianxian Zheng, Gregory Driessens, Bruno Gomes, Manfred Kraus, Chunxiao Xu, Yanping Zhang, Giorgio Kradjian, Guozhong Qin, Jin Qi, Xiaomei Xu, Bo Marelli, Huakui Yu, Wilson Guzman, Rober Tighe, Rachel Salazar, Kin-Ming Lo, Jessie English, Laszlo Radvanyi, Yan Lan, Michael Postow, Yasin Senbabaoglu, Billel Gasmi, Hong Zhong, Cailian Liu, Daniel Hirschhorhn-Cymerman, Yuanyuan Zha, Gregory Malnassy, Noreen Fulton, Jae-Hyun Park, Wendy Stock, Yusuke Nakamura, Hongtao Liu, Xiaoming Ju, Rachelle Kosoff, Kimberly Ramos, Brandon Coder, Robert Petit, Michael Princiotta, Kyle Perry, Jun Zou, Ainhoa Arina, Christian Fernandez, Wenxin Zheng, Michael A. Beckett, Helena J. Mauceri, Yang-Xin Fu, Ralph R. Weichselbaum, Whitney Lewis, Yanyan Han, Yeting Wu, Chou Yang, Jing Huang, Dongyun Wu, Jin Li, Xiaoling Liang, Xiangjun Zhou, Jinlin Hou, Raffit Hassan, Thierry Jahan, Scott J. Antonia, Hedy L. Kindler, Evan W. Alley, Somayeh Honarmand, Weiqun Liu, Meredith L. Leong, Chan C. Whiting, Nitya Nair, Amanda Enstrom, Edward E. Lemmens, Takahiro Tsujikawa, Sushil Kumar, Lisa M. Coussens, Aimee L. Murphy, Dirk G. Brockstedt, Sven D. Koch, Martin Sebastian, Christian Weiss, Martin Früh, Miklos Pless, Richard Cathomas, Wolfgang Hilbe, Georg Pall, Thomas Wehler, Jürgen Alt, Helge Bischoff, Michael Geissler, Frank Griesinger, Jens Kollmeier, Alexandros Papachristofilou, Fatma Doener, Mariola Fotin-Mleczek, Madeleine Hipp, Henoch S. Hong, Karl-Josef Kallen, Ute Klinkhardt, Claudia Stosnach, Birgit Scheel, Andreas Schroeder, Tobias Seibel, Ulrike Gnad-Vogt, Alfred Zippelius, Ha-Ram Park, Yong-Oon Ahn, Tae Min Kim, Soyeon Kim, Seulki Kim, Yu Soo Lee, Bhumsuk Keam, Dong-Wan Kim, Dae Seog Heo, Shari Pilon-Thomas, Amy Weber, Jennifer Morse, Krithika Kodumudi, Hao Liu, John Mullinax, Amod A. Sarnaik, Luke Pike, Andrew Bang, Tracy Balboni, Allison Taylor, Alexander Spektor, Tyler Wilhite, Monica Krishnan, Daniel Cagney, Brian Alexander, Ayal Aizer, Elizabeth Buchbinder, Mark Awad, Leena Ghandi, Jonathan Schoenfeld, Elizabeth Lessey-Morillon, Lisa Ridnour, Neil H. Segal, Manish Sharma, Dung T. Le, Robert L. Ferris, Andrew D. Zelenetz, Ronald Levy, Izidore S. Lossos, Caron Jacobson, Radhakrishnan Ramchandren, John Godwin, A. Dimitrios Colevas, Roland Meier, Suba Krishnan, Xuemin Gu, Jaclyn Neely, John Timmerman, Claire I. Vanpouille-Box, Silvia C. Formenti, Sandra Demaria, Erik Wennerberg, Aranzazu Mediero, Bruce N. Cronstein, Michael P. Gustafson, AriCeli DiCostanzo, Courtney Wheatley, Chul-Ho Kim, Svetlana Bornschlegl, Dennis A. Gastineau, Bruce D. Johnson, Allan B. Dietz, Cameron MacDonald, Mark Bucsek, Guanxi Qiao, Bonnie Hylander, Elizabeth Repasky, William J. Turbitt, Yitong Xu, Andrea Mastro, Connie J. Rogers, Sita Withers, Ziming Wang, Lam T. Khuat, Cordelia Dunai, Bruce R. Blazar, Dan Longo, Robert Rebhun, Steven K. Grossenbacher, Arta Monjazeb, William J. Murphy, Scott Rowlinson, Giulia Agnello, Susan Alters, David Lowe, Nicole Scharping, Ashley V. Menk, Ryan Whetstone, Xue Zeng, Greg M. Delgoffe, Patricia M. Santos, Jian Shi, Greg Delgoffe, Misako Nagasaka, Ammar Sukari, Miranda Byrne-Steele, Wenjing Pan, Xiaohong Hou, Brittany Brown, Mary Eisenhower, Jian Han, Natalie Collins, Robert Manguso, Hans Pope, Yashaswi Shrestha, Jesse Boehm, W. Nicholas Haining, Kyle R. Cron, Ayelet Sivan, Keston Aquino-Michaels, Marco Orecchioni, Davide Bedognetti, Wouter Hendrickx, Claudia Fuoco, Filomena Spada, Francesco Sgarrella, Gianni Cesareni, Francesco Marincola, Kostas Kostarelos, Alberto Bianco, Lucia Delogu, Jessica Roelands, Sabri Boughorbel, Julie Decock, Scott Presnell, Ena Wang, Franco M. Marincola, Peter Kuppen, Michele Ceccarelli, Darawan Rinchai, Damien Chaussabel, Lance Miller, Andrew Nguyen, J. Zachary Sanborn, Charles Vaske, Shahrooz Rabizadeh, Kayvan Niazi, Steven Benz, Shashank Patel, Nicholas Restifo, James White, Sam Angiuoli, Mark Sausen, Sian Jones, Maria Sevdali, John Simmons, Victor Velculescu, Luis Diaz, Theresa Zhang, Jennifer S. Sims, Sunjay M. Barton, Angela Kadenhe-Chiweshe, Filemon Dela Cruz, Andrew T. Turk, Christopher F. Mazzeo, Andrew L. Kung, Jeffrey N. Bruce, Darrell J. Yamashiro, Eileen P. Connolly, Jason Baird, Marka Crittenden, David Friedman, Hong Xiao, Rom Leidner, Bryan Bell, Kristina Young, Michael Gough, Zhen Bian, Koby Kidder, Yuan Liu, Emily Curran, Xiufen Chen, Leticia P. Corrales, Justin Kline, Ethan G. Aguilar, Jennifer Guerriero, Alaba Sotayo, Holly Ponichtera, Alexandra Pourzia, Sara Schad, Ruben Carrasco, Suzan Lazo, Roderick Bronson, Anthony Letai, Richard S. Kornbluth, Sachin Gupta, James Termini, Elizabeth Guirado, Geoffrey W. Stone, Christina Meyer, Laura Helming, Nicholas Wilson, Robert Hofmeister, Natalie J. Neubert, Laure Tillé, David Barras, Charlotte Soneson, Petra Baumgaertner, Donata Rimoldi, David Gfeller, Mauro Delorenzi, Silvia A. Fuertes Marraco, Daniel E. Speiser, Tara S. Abraham, Bo Xiang, Michael S. Magee, Scott A. Waldman, Adam E. Snook, Wojciech Blogowski, Ewa Zuba-Surma, Marta Budkowska, Daria Salata, Barbara Dolegowska, Teresa Starzynska, Leo Chan, Srinivas Somanchi, Kelsey McCulley, Dean Lee, Nico Buettner, Feng Shi, Paisley T. Myers, Stuart Curbishley, Sarah A. Penny, Lora Steadman, David Millar, Ellen Speers, Nicola Ruth, Gabriel Wong, Robert Thimme, David Adams, Mark Cobbold, Remy Thomas, Mariam Al-Muftah, Michael KK Wong, Michael Morse, Joseph I. Clark, Howard L. Kaufman, Gregory A. Daniels, Hong Hua, Tharak Rao, Janice P. Dutcher, Kai Kang, Yogen Saunthararajah, Vamsidhar Velcheti, Vikas Kumar, Firoz Anwar, Amita Verma, Zinal Chheda, Gary Kohanbash, John Sidney, Kaori Okada, Shruti Shrivastav, Diego A. Carrera, Shuming Liu, Naznin Jahan, Sabine Mueller, Ian F. Pollack, Angel M. Carcaboso, Alessandro Sette, Yafei Hou, Hideho Okada, Jessica J. Field, Weiping Zeng, Vincent FS Shih, Che-Leung Law, Peter D. Senter, Shyra J. Gardai, Nicole M. Okeley, Jennifer G. Abelin, Abu Z. Saeed, Stacy A. Malaker, Jeffrey Shabanowitz, Stephen T. Ward, Donald F. Hunt, Pam Profusek, Laura Wood, Dale Shepard, Petros Grivas, Kerstin Kapp, Barbara Volz, Detlef Oswald, Burghardt Wittig, Manuel Schmidt, Julian P. Sefrin, Lars Hillringhaus, Valeria Lifke, Alexander Lifke, Anna Skaletskaya, Jose Ponte, Thomas Chittenden, Yulius Setiady, Eva Sivado, Vincent Thomas, Meddy El Alaoui, Sébastien Papot, Charles Dumontet, Mike Dyson, John McCafferty, Said El Alaoui, Praveen K. Bommareddy, Andrew Zloza, Frederick Kohlhapp, Ann W. Silk, Sachin Jhawar, Tomas Paneque, Jenna Newman, Pedro Beltran, Felicia Cao, Bang-Xing Hong, Tania Rodriguez-Cruz, Xiao-Tong Song, Stephen Gottschalk, Hugo Calderon, Sam Illingworth, Alice Brown, Kerry Fisher, Len Seymour, Brian Champion, Emma Eriksson, Jessica Wenthe, Ann-Charlotte Hellström, Gabriella Paul-Wetterberg, Angelica Loskog, Ioanna Milenova, Magnus Ståhle, Justyna Jarblad-Leja, Gustav Ullenhag, Anna Dimberg, Rafael Moreno, Ramon Alemany, Sharad Goyal, Ann Silk, Janice Mehnert, Nashat Gabrail, Jennifer Bryan, Daniel Medina, Leah Mitchell, Kader Yagiz, Fernando Lopez, Daniel Mendoza, Anthony Munday, Harry Gruber, Douglas Jolly, Steven Fuhrmann, Sasa Radoja, Wei Tan, Aldo Pourchet, Alan Frey, Ian Mohr, Matthew Mulvey, Robert H. I. Andtbacka, Merrick Ross, Sanjiv Agarwala, Kenneth Grossmann, Matthew Taylor, John Vetto, Rogerio Neves, Adil Daud, Hung Khong, Stephanie M. Meek, Richard Ungerleider, Scott Welden, Maki Tanaka, Matthew Williams, Sigrun Hallmeyer, Bernard Fox, Zipei Feng, Christopher Paustian, Carlo Bifulco, Sadia Zafar, Otto Hemminki, Simona Bramante, Lotta Vassilev, Hongjie Wang, Andre Lieber, Silvio Hemmi, Tanja de Gruijl, Anna Kanerva, Tameem Ansari, Srividya Sundararaman, Diana Roen, Paul Lehmann, Anja C. Bloom, Lewis H. Bender, Ian B. Walters, Jay A. Berzofsky, Fanny Chapelin, Eric T. Ahrens, Jeff DeFalco, Michael Harbell, Amy Manning-Bog, Alexander Scholz, Danhui Zhang, Gilson Baia, Yann Chong Tan, Jeremy Sokolove, Dongkyoon Kim, Kevin Williamson, Xiaomu Chen, Jillian Colrain, Gregg Espiritu Santo, Ngan Nguyen, Wayne Volkmuth, Norman Greenberg, William Robinson, Daniel Emerling, Charles G. Drake, Daniel P. Petrylak, Emmanuel S. Antonarakis, Adam S. Kibel, Nancy N. Chang, Tuyen Vu, Dwayne Campogan, Heather Haynes, James B. Trager, Nadeem A. Sheikh, David I. Quinn, Peter Kirk, Murali Addepalli, Thomas Chang, Ping Zhang, Marina Konakova, Katsunobu Hagihara, Steven Pai, Laurie VanderVeen, Palakshi Obalapur, Peiwen Kuo, Phi Quach, Lawrence Fong, Deborah H. Charych, Jonathan Zalevsky, John L. Langowski, Yolanda Kirksey, Ravi Nutakki, Shalini Kolarkar, Rhoneil Pena, Ute Hoch, Stephen K. Doberstein, John Cha, Zach Mallon, Myra Perez, Amanda McDaniel, Snjezana Anand, Darrin Uecker, Richard Nuccitelli, Eva Wieckowski, Ravikumar Muthuswamy, Roshni Ravindranathan, Ariana N. Renrick, Menaka Thounaojam, Portia Thomas, Samuel Pellom, Anil Shanker, Duafalia Dudimah, Alan Brooks, Yu-Lin Su, Tomasz Adamus, Qifang Zhang, Sergey Nechaev, Marcin Kortylewski, Spencer Wei, Clark Anderson, Chad Tang, Jonathan Schoenhals, Efrosini Tsouko, John Heymach, Patricia de Groot, Joe Chang, Kenneth R. Hess, Adi Diab, Padmanee Sharma, David Hong, James Welsh, Andrea J. Parsons, Jardin Leleux, Stephane Ascarateil, Marie Eve Koziol, Dina Bai, Peihong Dai, Weiyi Wang, Ning Yang, Stewart Shuman, Liang Deng, Patrick Dillon, Gina Petroni, David Brenin, Kim Bullock, Walter Olson, Mark E. Smolkin, Kelly Smith, Carmel Nail, Craig L. Slingluff, Meenu Sharma, Faisal Fa’ak, Louise Janssen, Hiep Khong, Zhilan Xiao, Yared Hailemichael, Manisha Singh, Christina Vianden, Willem W. Overwijk, Andrea Facciabene, Pierini Stefano, Fang Chongyung, Stavros Rafail, Michael Nielsen, Peter Vanderslice, Darren G. Woodside, Robert V. Market, Ronald J. Biediger, Upendra K. Marathi, Kevin Hollevoet, Nick Geukens, Paul Declerck, Nathalie Joly, Laura McIntosh, Eustache Paramithiotis, Magnus Rizell, Malin Sternby, Bengt Andersson, Alex Karlsson-Parra, Rui Kuai, Lukasz Ochyl, Anna Schwendeman, James Moon, Weiwen Deng, Thomas E. Hudson, Bill Hanson, Chris S. Rae, Joel Burrill, Justin Skoble, George Katibah, Michele deVries, Peter Lauer, Thomas W. Dubensky, Xin Chen, Li Zhou, Xiubao Ren, Charu Aggarwal, Drishty Mangrolia, Roger Cohen, Gregory Weinstein, Matthew Morrow, Joshua Bauml, Kim Kraynyak, Jean Boyer, Jian Yan, Jessica Lee, Laurent Humeau, Sandra Oyola, Susan Duff, David Weiner, Zane Yang, Mark Bagarazzi, Douglas G. McNeel, Jens Eickhoff, Robert Jeraj, Mary Jane Staab, Jane Straus, Brian Rekoske, Glenn Liu, Marit Melssen, William Grosh, Nikole Varhegyi, Nadejda Galeassi, Donna H. Deacon, Elizabeth Gaughan, Maurizio Ghisoli, Minal Barve, Robert Mennel, Gladice Wallraven, Luisa Manning, Neil Senzer, John Nemunaitis, Masahiro Ogasawara, Shuichi Ota, Kaitlin M. Peace, Diane F. Hale, Timothy J. Vreeland, Doreen O. Jackson, John S. Berry, Alfred F. Trappey, Garth S. Herbert, Guy T. Clifton, Mark O. Hardin, Anne Toms, Na Qiao, Jennifer Litton, George E. Peoples, Elizabeth A. Mittendorf, Lila Ghamsari, Emilio Flano, Judy Jacques, Biao Liu, Jonathan Havel, Vladimir Makarov, Timothy A. Chan, Jessica B. Flechtner, John Facciponte, Stefano Ugel, Francesco De Sanctis, George Coukos, Sébastien Paris, Agnes Pottier, Laurent Levy, Bo Lu, Federica Cappuccini, Emily Pollock, Richard Bryant, Freddie Hamdy, Adrian Hill, Irina Redchenko, Hussein Sultan, Takumi Kumai, Valentyna Fesenkova, Esteban Celis, Ingrid Fernando, Claudia Palena, Justin M. David, Elizabeth Gabitzsch, Frank Jones, James L. Gulley, Mireia Uribe Herranz, Hiroshi Wada, Atsushi Shimizu, Toshihiro Osada, Satoshi Fukaya, Eiji Sasaki, Milad Abolhalaj, David Askmyr, Kristina Lundberg, Ann-Sofie Albrekt, Lennart Greiff, Malin Lindstedt, Dallas B. Flies, Tomoe Higuchi, Wojciech Ornatowski, Jaryse Harris, Sarah F. Adams, Todd Aguilera, Marjan Rafat, Laura Castellini, Hussein Shehade, Mihalis Kariolis, Dadi Jang, Rie vonEbyen, Edward Graves, Lesley Ellies, Erinn Rankin, Albert Koong, Amato Giaccia, Reham Ajina, Shangzi Wang, Jill Smith, Mariaelena Pierobon, Sandra Jablonski, Emanuel Petricoin, Louis M. Weiner, Lorcan Sherry, John Waller, Mark Anderson, Alison Bigley, Chantale Bernatchez, Cara Haymaker, Harriet Kluger, Michael Tetzlaff, Natalie Jackson, Ivan Gergel, Mary Tagliaferri, Patrick Hwu, Mario Snzol, Michael Hurwitz, Theresa Barberi, Allison Martin, Rahul Suresh, David Barakat, Sarah Harris-Bookman, Charles Drake, Alan Friedman, Sara Berkey, Stephanie Downs-Canner, Robert P. Edwards, Tyler Curiel, Kunle Odunsi, Tullia C. Bruno, Brandon Moore, Olivia Squalls, Peggy Ebner, Katherine Waugh, John Mitchell, Wilbur Franklin, Daniel Merrick, Martin McCarter, Brent Palmer, Jeffrey Kern, Dario Vignali, Jill Slansky, Anissa S. H. Chan, Xiaohong Qiu, Kathryn Fraser, Adria Jonas, Nadine Ottoson, Keith Gordon, Takashi O. Kangas, Steven Leonardo, Kathleen Ertelt, Richard Walsh, Mark Uhlik, Jeremy Graff, Nandita Bose, Ravi Gupta, Nitin Mandloi, Kiran Paul, Ashwini Patil, Rekha Sathian, Aparna Mohan, Malini Manoharan, Amitabha Chaudhuri, Yu Chen, Jing Lin, Yun-bin Ye, Chun-wei Xu, Gang Chen, Zeng-qing Guo, Andrey Komarov, Alex Chenchik, Michael Makhanov, Costa Frangou, Yi Zheng, Carla Coltharp, Darryn Unfricht, Ryan Dilworth, Leticia Fridman, Linying Liu, Milind Rajopadhye, Peter Miller, Fernando Concha-Benavente, Julie Bauman, Sumita Trivedi, Raghvendra Srivastava, James Ohr, Dwight Heron, Uma Duvvuri, Seungwon Kim, Heather Torrey, Toshi Mera, Yoshiaki Okubo, Eva Vanamee, Rosemary Foster, Denise Faustman, Edward Stack, Daisuke Izaki, Kristen Beck, Dan Tong Jia, Paul Armenta, Ashley White-Stern, Douglas Marks, Bret Taback, Basil Horst, Laura Hix Glickman, David B. Kanne, Kelsey S. Gauthier, Anthony L. Desbien, Brian Francica, Justin L. Leong, Leonard Sung, Ken Metchette, Shailaja Kasibhatla, Anne Marie Pferdekamper, Lianxing Zheng, Charles Cho, Yan Feng, Jeffery M. McKenna, John Tallarico, Steven Bender, Chudi Ndubaku, Sarah M. McWhirter, Elena Gonzalez Gugel, Charles J. M. Bell, Adiel Munk, Luciana Muniz, Nina Bhardwaj, Fei Zhao, Kathy Evans, Christine Xiao, Alisha Holtzhausen, Brent A. Hanks, Nathalie Scholler, Catherine Yin, Pien Van der Meijs, Andrew M. Prantner, Cecile M. Krejsa, Leia Smith, Brian Johnson, Daniel Branstetter, Paul L. Stein, Juan C. Jaen, Joanne BL Tan, Ada Chen, Timothy Park, Jay P. Powers, Holly Sexton, Guifen Xu, Steve W. Young, Ulrike Schindler, Wentao Deng, David John Klinke, Hannah M. Komar, Gregory Serpa, Omar Elnaggar, Philip Hart, Carl Schmidt, Mary Dillhoff, Ming Jin, Michael C. Ostrowski, Madhuri Koti, Katrina Au, Nichole Peterson, Peter Truesdell, Gillian Reid-Schachter, Charles Graham, Andrew Craig, Julie-Ann Francis, Beatrix Kotlan, Timea Balatoni, Emil Farkas, Laszlo Toth, Mihaly Ujhelyi, Akos Savolt, Zoltan Doleschall, Szabolcs Horvath, Klara Eles, Judit Olasz, Orsolya Csuka, Miklos Kasler, Gabriella Liszkay, Eytan Barnea, Collin Blakely, Patrick Flynn, Reid Goodman, Raphael Bueno, David Sugarbaker, David Jablons, V. Courtney Broaddus, Brian West, Paul R. Kunk, Joseph M. Obeid, Kevin Winters, Patcharin Pramoonjago, Edward B. Stelow, Todd W. Bauer, Osama E. Rahma, Adam Lamble, Yoko Kosaka, Fei Huang, Kate A. Saser, Homer Adams, Christina E. Tognon, Ted Laderas, Shannon McWeeney, Marc Loriaux, Jeffery W. Tyner, Brian J. Druker, Evan F. Lind, Zhuqing Liu, Shanhong Lu, Lawrence P. Kane, Gulidanna Shayan, Julia Femel, Ryan Lane, Jamie Booth, Amanda W. Lund, Anthony Rodriguez, Victor H. Engelhard, Alessandra Metelli, Bill X. Wu, Caroline W. Fugle, Rachidi Saleh, Shaoli Sun, Jennifer Wu, Bei Liu, Zihai Li, Zachary S. Morris, Emily I. Guy, Clinton Heinze, Jasdeep Kler, Monica M. Gressett, Lauryn R. Werner, Stephen D. Gillies, Alan J. Korman, Hans Loibner, Jacquelyn A. Hank, Alexander L. Rakhmilevich, Paul M. Harari, Paul M. Sondel, Erica Huelsmann, Joseph Broucek, Dorothee Brech, Tobias Straub, Martin Irmler, Johannes Beckers, Florian Buettner, Elke Schaeffeler, Matthias Schwab, Elfriede Noessner, Alison Wolfreys, Andre Da Costa, John Silva, Andrea Crosby, Ludovicus Staelens, Graham Craggs, Annick Cauvin, Sean Mason, Alison M. Paterson, Andrew C. Lake, Caroline M. Armet, Rachel W. O’Connor, Jonathan A. Hill, Emmanuel Normant, Ammar Adam, Detlev M. Biniszkiewicz, Scott C. Chappel, Vito J. Palombella, Pamela M. Holland, Annette Becker, Manmohan R. Leleti, Eric Newcomb, Joanne B. L. Tan, Suthee Rapisuwon, Arash Radfar, Kellie Gardner, Geoffrey Gibney, Michael Atkins, Keith R. Rennier, Robert Crowder, Ping Wang, Russell K. Pachynski, Rosa M. Santana Carrero, Sarai Rivas, Figen Beceren-Braun, Scott Anthony, Kimberly S. Schluns, Deepali Sawant, Maria Chikina, Hiroshi Yano, Creg Workman, Elise Salerno, Ileana Mauldin, Donna Deacon, Sofia Shea, Joel Pinczewski, Thomas Gajewski, Stefani Spranger, Brendan Horton, Akiko Suzuki, Pamela Leland, Bharat H. Joshi, Raj K. Puri, Randy F. Sweis, Riyue Bao, Jason Luke, Marie-Nicole Theodoraki, Frances-Mary Mogundo, Haejung Won, Dayson Moreira, Chan Gao, Xingli Zhao, Priyanka Duttagupta, Jeremy Jones, Massimo D’Apuzzo, and Sumanta Pal
- Subjects
0301 basic medicine ,Pharmacology ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Immunology ,Cancer ,Immunotherapy ,medicine.disease ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Family medicine ,Molecular Medicine ,Immunology and Allergy ,Medicine ,business - Abstract
O1 IL-15 primes an mTOR-regulated gene-expression program to prolong anti-tumor capacity of human natural killer cells #### Andreas Lundqvist1, Vincent van Hoef1, Xiaonan Zhang1, Erik Wennerberg2, Julie Lorent1, Kristina Witt1, Laia Masvidal Sanz1, Shuo Liang1, Shannon Murray3, Ola Larsson1
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.