68 results on '"Piers E.M. Patten"'
Search Results
2. Targeting the tumor microenvironment in chronic lymphocytic leukemia
- Author
-
Rebecka Svanberg, Sine Janum, Piers E.M. Patten, Alan G. Ramsay, and Carsten U. Niemann
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
The tumor microenvironment (TME) plays an essential role in the development, growth, and survival of the malignant B-cell clone in chronic lymphocytic leukemia (CLL). Within the proliferation niches of lymph nodes, bone marrow, and secondary lymphoid organs, a variety of phenotypically and functionally altered cell types, including T cells, natural killer cells, monocytes/macrophages, endothelial and mesenchymal stroma cells, provide crucial survival signals, along with CLL-cellinduced suppression of antitumor immune responses. The B-cell receptor pathway plays a pivotal role in mediating the interaction between CLL cells and the TME. However, an increasing number of additional components of the multifactorial TME are being discovered. Although the majority of therapeutic strategies employed in CLL hitherto have focused on targeting the leukemic cells, emerging evidence implies that modulation of microenvironmental cells and CLL-TME interactions by novel therapeutic agents significantly affect their clinical efficacy. Thus, improving our understanding of CLL-TME interactions and how they are affected by current therapeutic agents may improve and guide treatment strategies. Identification of novel TME interactions may also pave the road for the development of novel therapeutic strategies targeting the TME. In this review, we summarize current evidence on the effects of therapeutic agents on cells and interactions within the TME. With a growing demand for improved and personalized treatment options in CLL, this review aims at inspiring future exploration of smart drug combination strategies, translational studies, and novel therapeutic targets in clinical trials.
- Published
- 2021
- Full Text
- View/download PDF
3. The architecture of neoplastic follicles in follicular lymphoma; analysis of the relationship between the tumor and follicular helper T cells
- Author
-
William Townsend, Marta Pasikowska, Deborah Yallop, Elizabeth H. Phillips, Piers E.M. Patten, Jonathan R. Salisbury, Robert Marcus, Andrea Pepper, and Stephen Devereux
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
CD4+ T-follicular helper cells are essential for the survival, proliferation, and differentiation of germinal center B cells and have been implicated in the pathogenesis of follicular lymphoma (FL). To further define the role of these cells in FL, we used multiparameter confocal microscopy to compare the architecture of normal and neoplastic follicles and next generation sequencing to analyze the T-cell receptor repertoire in FL lymph nodes (LN). Multiparameter analysis of LN showed that the proportion of T-follic-ular helper cells (TFH) in normal and neoplastic follicles is the same and that the previously reported increase in TFH numbers in FL is thus due to an increase in the number and not content of follicles. As in normal germinal centers, TFH were shown to have a close spatial correlation with proliferating B cells in neoplastic follicles, where features of immunological synapse formation were observed. The number of TFH in FL correlate with the rate of B-cell proliferation and TFH co-localized to activation induced cytidine deaminase expressing proliferating B cells. T-cell receptor repertoire analysis of FL LN revealed that follicular areas are significantly more clonal when compared to the rest of the LN. These novel findings show that neoplastic follicles and germinal centers share important structural features and provide further evidence that TFH may play a role in driving B-cell proliferation and genomic evolution in TFH. Our results also suggest that targeting this interaction would be an attractive therapeutic option.
- Published
- 2020
- Full Text
- View/download PDF
4. Immunogenicity of SARS-CoV-2 vaccines in patients with cancer
- Author
-
Helen Kakkassery, Esme Carpenter, Piers E.M. Patten, and Sheeba Irshad
- Subjects
COVID-19 Vaccines ,Ad26COVS1 ,SARS-CoV-2 ,ChAdOx1 nCoV-19 ,Neoplasms ,Humans ,COVID-19 ,Molecular Medicine ,Viral Vaccines ,Molecular Biology ,BNT162 Vaccine - Abstract
Transmission of the SARS-CoV-2 virus and its corresponding disease (COVID-19) has been shown to impose a higher burden on cancer patients than on the general population. Approved vaccines for use include new technology mRNA vaccines such as BNT162b2 (Pfizer-BioNTech) and mRNA-1273 (Moderna), and nonreplicating viral vector vaccines such as Ad26.COV2.S (JohnsonJohnson) and AZD1222 (AstraZeneca). Impaired or delayed humoral and diminished T-cell responses are evident in patients with cancer, especially in patients with haematological cancers or those under active chemotherapy. Herein we review the current data on vaccine immunogenicity in cancer patients, including recommendations for current practice and future research.
- Published
- 2022
5. Long-Term Clinical Outcomes and Correlative Efficacy Analyses in Patients (Pts) with Relapsed/Refractory Follicular Lymphoma (r/r FL) Treated with Tisagenlecleucel in the Elara Trial
- Author
-
Martin Dreyling, Michael Dickinson, Joaquin Martinez Lopez, Arne Kolstad, Jason P Butler, Monalisa Ghosh, Leslie L. Popplewell, Julio Chavez, Emmanuel Bachy, Koji Kato, Hideo Harigae, Marie Jose Kersten, Charalambos Andreadis, Peter A. Riedell, Phoebe Joy Ho, Jose A. Perez-Simon, Andy Chen, Loretta J. Nastoupil, Bastian von Tresckow, Andrés J M Ferreri, Takanori Teshima, Piers E.M. Patten, Joseph P. McGuirk, Andreas Petzer, Fritz Offner, Andreas Viardot, Pier Luigi Zinzani, Ram Malladi, Ines Paule, Aiesha Zia, Rakesh Awasthi, Xia Han, Davide Germano, Darragh O'Donovan, Roberto Ramos, Aisha Masood, Catherine Thieblemont, Nathan H. Fowler, and Stephen J. Schuster
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
6. Supplementary Figure 1 from Interaction with Vascular Endothelium Enhances Survival in Primary Chronic Lymphocytic Leukemia Cells via NF-κB Activation and De novo Gene Transcription
- Author
-
Stephen Devereux, Chris Fegan, Ghulam J. Mufti, N. Shaun B. Thomas, Deborah Yallop, Najeem'deen Folarin, Jane Moorhead, Satyen Gohil, Saman Hewamana, Piers E.M. Patten, Chris Pepper, and Andrea G.S. Buggins
- Abstract
Supplementary Figure 1 from Interaction with Vascular Endothelium Enhances Survival in Primary Chronic Lymphocytic Leukemia Cells via NF-κB Activation and De novo Gene Transcription
- Published
- 2023
7. Supplementary Table 2 from Interaction with Vascular Endothelium Enhances Survival in Primary Chronic Lymphocytic Leukemia Cells via NF-κB Activation and De novo Gene Transcription
- Author
-
Stephen Devereux, Chris Fegan, Ghulam J. Mufti, N. Shaun B. Thomas, Deborah Yallop, Najeem'deen Folarin, Jane Moorhead, Satyen Gohil, Saman Hewamana, Piers E.M. Patten, Chris Pepper, and Andrea G.S. Buggins
- Abstract
Supplementary Table 2 from Interaction with Vascular Endothelium Enhances Survival in Primary Chronic Lymphocytic Leukemia Cells via NF-κB Activation and De novo Gene Transcription
- Published
- 2023
8. Supplementary Figure 2 from Interaction with Vascular Endothelium Enhances Survival in Primary Chronic Lymphocytic Leukemia Cells via NF-κB Activation and De novo Gene Transcription
- Author
-
Stephen Devereux, Chris Fegan, Ghulam J. Mufti, N. Shaun B. Thomas, Deborah Yallop, Najeem'deen Folarin, Jane Moorhead, Satyen Gohil, Saman Hewamana, Piers E.M. Patten, Chris Pepper, and Andrea G.S. Buggins
- Abstract
Supplementary Figure 2 from Interaction with Vascular Endothelium Enhances Survival in Primary Chronic Lymphocytic Leukemia Cells via NF-κB Activation and De novo Gene Transcription
- Published
- 2023
9. Supplementary Table 1 from Interaction with Vascular Endothelium Enhances Survival in Primary Chronic Lymphocytic Leukemia Cells via NF-κB Activation and De novo Gene Transcription
- Author
-
Stephen Devereux, Chris Fegan, Ghulam J. Mufti, N. Shaun B. Thomas, Deborah Yallop, Najeem'deen Folarin, Jane Moorhead, Satyen Gohil, Saman Hewamana, Piers E.M. Patten, Chris Pepper, and Andrea G.S. Buggins
- Abstract
Supplementary Table 1 from Interaction with Vascular Endothelium Enhances Survival in Primary Chronic Lymphocytic Leukemia Cells via NF-κB Activation and De novo Gene Transcription
- Published
- 2023
10. Combination of Ibrutinib Plus Venetoclax with MRD-Driven Duration of Treatment Results in a Higher Rate of MRD Negativity in IGHV Unmutated Than Mutated CLL: Updated Interim Analysis of FLAIR Study
- Author
-
Talha Munir, Alexandra Pitchford, Adrian Bloor, Andrew Pettitt, Piers E.M. Patten, Francesco Forconi, Anna Schuh, Christopher P Fox, Simona Gatto, Ben Kennedy, John Gribben, Nicholas Pemberton, Oonagh Sheehy, Gavin Preston, Dena Howard, Anna Hockaday, David Cairns, Sharon Jackson, Natasha Greatorex, Nichola McWhirter, Jane Shingles, Kate Cwynarski, Shankara Paneesha, David Allsup, Andrew Rawstron, and Peter Hillmen
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
11. Richter transformation of chronic lymphocytic leukaemia: a British Society for Haematology Good Practice Paper
- Author
-
Toby A. Eyre, John Riches, Peter Hillmen, George A Follows, Piers E.M. Patten, Renata Walewska, Helen Marr, and Anna Schuh
- Subjects
Male ,medicine.medical_specialty ,Hematology ,Lymphocytic leukaemia ,Richter transformation ,business.industry ,Prognosis ,Leukemia, Lymphocytic, Chronic, B-Cell ,Dermatology ,England ,Internal medicine ,medicine ,Humans ,Female ,business ,Good practice - Published
- 2021
12. Management of cardiovascular complications of bruton tyrosine kinase inhibitors
- Author
-
Renata Walewska, Nilima Parry-Jones, Sunil Iyengar, Anna Schuh, Alexander R. Lyon, Terry McCormack, Piers E.M. Patten, Peter Hillmen, Gregory Y.H. Lip, Nicolas Martinez-Calle, and Chloe Pek Sang Tang
- Subjects
medicine.medical_specialty ,hypertension ,Cardiovascular Complication ,Clinical Decision-Making ,Cardiovascular System ,sudden cardiac death ,Sudden cardiac death ,Diagnosis, Differential ,chemistry.chemical_compound ,Risk Factors ,bruton tyrosine kinase inhibitor ,ibrutinib ,Internal medicine ,Agammaglobulinaemia Tyrosine Kinase ,medicine ,Animals ,Humans ,Bruton's tyrosine kinase ,atrial fibrillation ,Protein Kinase Inhibitors ,cardiovascular complication ,biology ,business.industry ,Disease Management ,Atrial fibrillation ,Hematology ,medicine.disease ,chemistry ,Cardiovascular Diseases ,Ibrutinib ,biology.protein ,Cardiology ,Disease Susceptibility ,business - Published
- 2021
13. Risk of COVID-19 death in cancer patients: an analysis from Guy’s Cancer Centre and King’s College Hospital in London
- Author
-
Richard Sullivan, Saoirse Dolly, Gincy George, Piers E.M. Patten, Kieran Palmer, Elinor J. Sawyer, Andrea D'Souza, James Spicer, Simon Gomberg, Reuben Benjamin, Paul Ross, Kamarul Zaki, Anna Haire, Ailsa Sita-Lumsden, Austin G. Kulasekararaj, Sophie Papa, Fidelma Cahill, Muhammed Mansour Ceesay, Victoria Potter, Sheeba Irshad, Thinzar Ko Ko, Mieke Van Hemelrijck, Beth Russell, Debra H. Josephs, Claire N. Harrison, Paul Fields, Danielle Crawley, Antonio Pagliuca, Vallari Shah, Deborah Enting, Rushan Sylva, Angela Swampillai, Harriet Wylie, Charlotte Moss, Anne Rigg, David Wrench, Shahram Kordasti, and Maria J. Monroy-Iglesias
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Patient characteristics ,Article ,Internal medicine ,Neoplasms ,Cox proportional hazards regression ,Cancer centre ,London ,medicine ,Humans ,Aged ,Aged, 80 and over ,business.industry ,SARS-CoV-2 ,Cancer ,COVID-19 ,Translational research ,Middle Aged ,medicine.disease ,Hospitals ,Risk factors ,Oncology ,Raised CRP ,Hematologic Neoplasms ,Haematological cancer ,Female ,business - Abstract
Background Using an updated dataset with more patients and extended follow-up, we further established cancer patient characteristics associated with COVID-19 death. Methods Data on all cancer patients with a positive reverse transcription-polymerase chain reaction swab for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) at Guy’s Cancer Centre and King’s College Hospital between 29 February and 31 July 2020 was used. Cox proportional hazards regression was performed to identify which factors were associated with COVID-19 mortality. Results Three hundred and six SARS-CoV-2-positive cancer patients were included. Seventy-one had mild/moderate and 29% had severe COVID-19. Seventy-two patients died of COVID-19 (24%), of whom 35 died 2–5 years [2.81 (1.41–5.59)] or ≥5 years were associated with an increased mortality. Age >60 years and raised C-reactive protein (CRP) were also associated with COVID-19 death. Haematological cancer, a longer-established cancer diagnosis, dyspnoea at diagnosis and raised CRP were indicative of early COVID-19-related death in cancer patients ( Conclusions Findings further substantiate evidence for increased risk of COVID-19 mortality for male and Asian cancer patients, and those with haematological malignancies or a cancer diagnosis >2 years. These factors should be accounted for when making clinical decisions for cancer patients.
- Published
- 2021
14. Safety and immunogenicity of one versus two doses of the COVID-19 vaccine BNT162b2 for patients with cancer: interim analysis of a prospective observational study
- Author
-
Jennifer Vidler, Shubhankar Sinha, Helen Kakkassery, Jack Cooper, Thanussuyah Alaguthurai, Thomas Hayday, Ana Montes, Yin Wu, Puay Ling Lee, Shraddha Kamdar, Leticia Monin, Muhammad Shamim Khan, Rosalind Graham, Daniel Davies, Maria Conde Poole, Miguel Muñoz-Ruiz, Adam Laing, Charlotte O'Brien-Gore, Sheeba Irshad, Sophie Papa, Anne Rigg, Elizabeth Harvey-Jones, Piers E.M. Patten, Magdalene Joseph, Paul Fields, Clara Domingo-Vila, Mark Harries, Francesca Di Rosa, Jeffrey Seow, Richard Davis, Adrian Hayday, Katie J. Doores, Duncan R. McKenzie, Isaac Francos Quijorna, Josephine Eum, Angela Swampillai, Timothy Tree, Bernard V. North, Liane Dupont, James Spicer, Michael H. Malim, Sultan Abdul-Jawad, Carl Graham, and Irene del Molino del Barrio
- Subjects
0301 basic medicine ,medicine.medical_specialty ,03 medical and health sciences ,0302 clinical medicine ,vaccine ,Internal medicine ,medicine ,cancer ,Seroconversion ,Prospective cohort study ,biology ,business.industry ,Immunogenicity ,COVID-19 ,Cancer ,Articles ,medicine.disease ,Interim analysis ,Vaccination ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Antibody ,business ,Blood sampling - Abstract
Summary Background The efficacy and safety profiles of vaccines against SARS-CoV-2 in patients with cancer is unknown. We aimed to assess the safety and immunogenicity of the BNT162b2 (Pfizer–BioNTech) vaccine in patients with cancer. Methods For this prospective observational study, we recruited patients with cancer and healthy controls (mostly health-care workers) from three London hospitals between Dec 8, 2020, and Feb 18, 2021. Participants who were vaccinated between Dec 8 and Dec 29, 2020, received two 30 μg doses of BNT162b2 administered intramuscularly 21 days apart; patients vaccinated after this date received only one 30 μg dose with a planned follow-up boost at 12 weeks. Blood samples were taken before vaccination and at 3 weeks and 5 weeks after the first vaccination. Where possible, serial nasopharyngeal real-time RT-PCR (rRT-PCR) swab tests were done every 10 days or in cases of symptomatic COVID-19. The coprimary endpoints were seroconversion to SARS-CoV-2 spike (S) protein in patients with cancer following the first vaccination with the BNT162b2 vaccine and the effect of vaccine boosting after 21 days on seroconversion. All participants with available data were included in the safety and immunogenicity analyses. Ongoing follow-up is underway for further blood sampling after the delayed (12-week) vaccine boost. This study is registered with the NHS Health Research Authority and Health and Care Research Wales (REC ID 20/HRA/2031). Findings 151 patients with cancer (95 patients with solid cancer and 56 patients with haematological cancer) and 54 healthy controls were enrolled. For this interim data analysis of the safety and immunogenicity of vaccinated patients with cancer, samples and data obtained up to March 19, 2021, were analysed. After exclusion of 17 patients who had been exposed to SARS-CoV-2 (detected by either antibody seroconversion or a positive rRT-PCR COVID-19 swab test) from the immunogenicity analysis, the proportion of positive anti-S IgG titres at approximately 21 days following a single vaccine inoculum across the three cohorts were 32 (94%; 95% CI 81–98) of 34 healthy controls; 21 (38%; 26–51) of 56 patients with solid cancer, and eight (18%; 10–32) of 44 patients with haematological cancer. 16 healthy controls, 25 patients with solid cancer, and six patients with haematological cancer received a second dose on day 21. Of the patients with available blood samples 2 weeks following a 21-day vaccine boost, and excluding 17 participants with evidence of previous natural SARS-CoV-2 exposure, 18 (95%; 95% CI 75–99) of 19 patients with solid cancer, 12 (100%; 76–100) of 12 healthy controls, and three (60%; 23–88) of five patients with haematological cancers were seropositive, compared with ten (30%; 17–47) of 33, 18 (86%; 65–95) of 21, and four (11%; 4–25) of 36, respectively, who did not receive a boost. The vaccine was well tolerated; no toxicities were reported in 75 (54%) of 140 patients with cancer following the first dose of BNT162b2, and in 22 (71%) of 31 patients with cancer following the second dose. Similarly, no toxicities were reported in 15 (38%) of 40 healthy controls after the first dose and in five (31%) of 16 after the second dose. Injection-site pain within 7 days following the first dose was the most commonly reported local reaction (23 [35%] of 65 patients with cancer; 12 [48%] of 25 healthy controls). No vaccine-related deaths were reported. Interpretation In patients with cancer, one dose of the BNT162b2 vaccine yields poor efficacy. Immunogenicity increased significantly in patients with solid cancer within 2 weeks of a vaccine boost at day 21 after the first dose. These data support prioritisation of patients with cancer for an early (day 21) second dose of the BNT162b2 vaccine. Funding King's College London, Cancer Research UK, Wellcome Trust, Rosetrees Trust, and Francis Crick Institute.
- Published
- 2021
15. Triggering interferon signaling in T cells with avadomide sensitizes CLL to anti-PD-L1/PD-1 immunotherapy
- Author
-
Jeffrey A. Jones, Piers E.M. Patten, Richard Rosenquist, Hsiling Chiu, Antonia Lopez-Girona, Benedetta Apollonio, Patrick Hagner, Martin S. Tallman, Matt Stokes, Farrukh T. Awan, Neil E. Kay, Anita Gandhi, Kostas Stamatopoulos, Anna Vardi, Rose-Marie Amini, Despoina Papazoglou, Alan G. Ramsay, Mariam Fanous, Nikolaos Ioannou, Preethi Janardhanan, Lesley-Ann Sutton, and Tait D. Shanafelt
- Subjects
Chemokine ,Combination therapy ,T-Lymphocytes ,medicine.medical_treatment ,Immunology ,Antineoplastic Agents ,Lymphocyte Activation ,Biochemistry ,Mice ,Interferon ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Immune Checkpoint Inhibitors ,Piperidones ,Quinazolinones ,Tumor microenvironment ,biology ,business.industry ,Cereblon ,Cell Biology ,Hematology ,Immunotherapy ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Xenograft Model Antitumor Assays ,Leukemia ,biology.protein ,Cancer research ,Interferons ,business ,CD8 ,Signal Transduction ,medicine.drug - Abstract
Cancer treatment has been transformed by checkpoint blockade therapies, with the highest anti-tumor activity of anti-programmed death 1 (PD-1) antibody therapy seen in Hodgkin lymphoma. Disappointingly, response rates have been low in the non-Hodgkin lymphomas, with no activity seen in relapsed/refractory chronic lymphocytic leukemia (CLL) with PD-1 blockade. Thus, identifying more powerful combination therapy is required for these patients. Here, we preclinically demonstrate enhanced anti-CLL activity following combinational therapy with anti-PD-1 or anti-PD-1 ligand (PD-L1) and avadomide, a cereblon E3 ligase modulator (CELMoD). Avadomide induced type I and II interferon (IFN) signaling in patient T cells, triggering a feedforward cascade of reinvigorated T-cell responses. Immune modeling assays demonstrated that avadomide stimulated T-cell activation, chemokine expression, motility and lytic synapses with CLL cells, as well as IFN-inducible feedback inhibition through upregulation of PD-L1. Patient-derived xenograft tumors treated with avadomide were converted to CD8+ T cell-inflamed tumor microenvironments that responded to anti-PD-L1/PD-1-based combination therapy. Notably, clinical analyses showed increased PD-L1 expression on T cells, as well as intratumoral expression of chemokine signaling genes in B-cell malignancy patients receiving avadomide-based therapy. These data illustrate the importance of overcoming a low inflammatory T-cell state to successfully sensitize CLL to checkpoint blockade-based combination therapy.
- Published
- 2021
16. MRD4 Eradication at 6 Months and Early Clearance of MRD with Combination of Ibrutinib Plus Venetoclax Results in Sustained Clinical and MRD Responses: Exploratory Analysis of the Blood Cancer UK TAP Clarity Study
- Author
-
Talha Munir, Louise-Rae Cherrill, Nichola Webster, Surita Dalal, Rebecca H. Boucher, Chhaya Sankhalpara, Francesca Yates, Sonia Fox, Donald Macdonald, Christopher Fegan, Alison McCaig, Anna Schuh, Andrew Pettitt, John G. Gribben, Piers E.M. Patten, Stephen Devereux, Adrian Bloor, Christopher P Fox, Francesco Forconi, Andy C. Rawstron, and Peter Hillmen
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
17. Humoral and cellular immunity to delayed second dose of SARS-CoV-2 BNT162b2 mRNA vaccination in patients with cancer
- Author
-
Timothy Tree, Bernard V. North, Sophie Papa, K. Owczarczyk, Sultan Abdul-Jawad, Hartmut Kristeleit, Kamila Sychowska, Piers E.M. Patten, Sheeba Irshad, Clara Domingo-Vila, Catherine Tremain, Jennifer Vidler, Paul Fields, Thomas Lechmere, Emily Pollock, Rosalind Graham, Carl Graham, Jeffrey Seow, Leticia Monin, Thanussuyah Alaguthurai, Miguel Muñoz-Ruiz, Michael H. Malim, Jack Cooper, Katie J. Doores, Charalampos Gousis, Adrian Hayday, Duncan R. McKenzie, and Angela Swampillai
- Subjects
Cancer Research ,2019-20 coronavirus outbreak ,Cellular immunity ,COVID-19 Vaccines ,Time Factors ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,T-Lymphocytes ,Antibodies, Viral ,Article ,Immunogenicity, Vaccine ,Neoplasms ,medicine ,Humans ,In patient ,BNT162 Vaccine ,Immunization Schedule ,Messenger RNA ,Immunity, Cellular ,business.industry ,SARS-CoV-2 ,Vaccination ,Case-control study ,Cancer ,COVID-19 ,medicine.disease ,Virology ,Immunity, Humoral ,Treatment Outcome ,Oncology ,Case-Control Studies ,Immunoglobulin G ,Host-Pathogen Interactions ,Spike Glycoprotein, Coronavirus ,Cytokines ,business - Published
- 2021
18. RUNIMC: An R-based package for imaging mass cytometry data analysis and pipeline validation
- Author
-
Kenrick Ng, Rami Mustapha, Paul R. Barber, Tony Ng, Luigi Dolcetti, Gregory Weitsman, Piers E.M. Patten, Julie Nuo En Chan, Selvam Thavaraj, and Jinhai Deng
- Subjects
business.industry ,Computer science ,Pipeline (computing) ,Pattern recognition ,Mass cytometry ,Artificial intelligence ,Gold standard (test) ,business ,Nuclear staining ,Head and neck ,Random forest - Abstract
We propose a novel pipeline for the analysis of imaging mass cytometry data, comparing an unbiased approach, representing the actual gold standard, with a novel biased method. We made use of both synthetic/ controlled datasets as well as two datasets obtained from FFPE sections of follicular lymphoma, and head and neck patients, stained with a 14 and 29-markers panels respectively. The novel pipeline, denominated RUNIMC, has been completely developed in R and contained in a single package. The novelty resides in the ease with which multi-class random forest classifier can be used to classify image features, making the pathologist’s and expert classification pivotal, and the use of a random forest regression approach that permits a better detection of cell boundaries, and alleviates the necessity of relying on a perfect nuclear staining.
- Published
- 2021
19. Efficacy and Safety of Tisagenlecleucel in Adult Patients with Relapsed/Refractory Follicular Lymphoma: Interim Analysis of the Phase 2 Elara Trial
- Author
-
Piers E.M. Patten, Julio C. Chavez, P. Joy Ho, Monalisa Ghosh, Jason Butler, Bastian von Tresckow, Andreas L. Petzer, Arne Kolstad, Alessandra Forcina, Catherine Thieblemont, José A. Pérez-Simón, Sarah J. Nagle, Pier Luigi Zinzani, Lida Bubuteishvili Pacaud, Joseph McGuirk, Aiesha Zia, Charalambos Andreadis, Michael Dickinson, Stephen J. Schuster, Marie José Kersten, Andrés J.M. Ferreri, Hideo Harigae, Loretta J. Nastoupil, Peter A. Riedell, Martin Dreyling, Fritz Offner, Ram Malladi, Andreas Viardot, Takanori Teshima, Koji Kato, Leslie Popplewell, Emmanuel Bachy, Joaquin Martinez-Lopez, and Nathan Fowler
- Subjects
Oncology ,medicine.medical_specialty ,Adult patients ,business.industry ,Immunology ,Follicular lymphoma ,Cell Biology ,Hematology ,medicine.disease ,Interim analysis ,Biochemistry ,Internal medicine ,Relapsed refractory ,medicine ,business - Abstract
Background: Follicular lymphoma (FL) remains an incurable disease for most patients (pts), characterized by a relapsing and remitting pattern. For pts with relapsed/refractory (r/r) FL, treatment outcomes typically worsen with each subsequent line of therapy, highlighting an unmet need. Tisagenlecleucel (tisa-cel) has demonstrated clinical benefits and manageable safety in pediatric and young adult pts with r/r B-cell acute lymphoblastic leukemia and adult pts with r/r diffuse large B-cell lymphoma. In a prior phase (Ph) 2a study of 14 pts with r/r FL, 71% achieved durable complete remission with tisa-cel (Chong et al. ICML 2019). Here we present a planned interim analysis of ELARA, a Ph 2, international trial of tisa-cel in adults with r/r FL. Methods: Adults with histologically confirmed FL (grades [Gr] 1-3A) being r/r within 6 mo after second-/later-line therapy (including an anti-CD20 monoclonal antibody with an alkylating agent) or relapsed after autologous hematopoietic stem cell transplant (autoHSCT), and with ECOG performance score of 0-1 were eligible. Pts with histologic transformation, prior allogeneic HSCT, or active CNS involvement were excluded. All pts received lymphodepleting chemotherapy followed by tisa-cel infusion of 0.6-6×108 CAR-T cells (bridging therapy prior to infusion was permitted). Disease was reassessed prior to infusion for all pts who received bridging therapy to establish a new baseline. After infusion, disease assessments were performed every 3 mo. The primary endpoint was complete response rate (CRR) based on best response by central review (Lugano 2014 criteria). Pts evaluable for efficacy had measurable disease at infusion and ≥6 mo follow-up from infusion or discontinued early. Secondary endpoints included overall response rate (ORR), duration of response (DOR), progression-free survival (PFS), overall survival (OS), and safety. Results: As of May 26, 2020, 122 pts were screened, 98 were enrolled, 97 received tisa-cel (median follow-up, 6.5 mo), and 52 were evaluable for efficacy (median follow-up, 9.9 mo). At study entry, median age among treated pts was 57 y (range, 29-73), 66% of pts were male, 84% had stage III-IV disease, and 60% had a FLIPI score ≥3. The median number of prior lines of therapy was 4 (range, 2-13), including prior autoHSCT in 36%; 77% were refractory to the last prior treatment (75% ≥2 prior regimens), and 60% had disease progression within 2 y of initial anti-CD20-containing treatment. Overall, 43% of pts received bridging therapy; 18% were treated as outpatient. Of the first 52 pts evaluable for efficacy, CRR was 65.4% (34/52; 99.5% CI, 45.1-82.4) in the intent-to-treat (ITT) population and 71.1% (33/46; 99.5% CI, 56.5-84.0) in the per-protocol (PP) population; ORR was 82.7% (43/52; 95% CI, 69.7-91.8) in the ITT population and 84.8% (39/46; 95% CI, 71.1-93.7) in the PP population. In pts with best response of CR, probability of response lasting ≥6 mo was 89.7% (84.4% for all responding pts [CR+PR]). CRR and ORR were consistent across key prognostic subgroups and per investigator assessment. Median DOR, PFS, OS, and time to next antilymphoma treatment were not reached. Of 97 pts evaluable for safety, 69% experienced Gr ≥3 adverse events, most commonly neutropenia (Gr 3, 13%; Gr 4, 15%); 48% of pts had cytokine release syndrome related to tisa-cel (CRS; Gr 1, 29%; Gr 2, 20%; Gr ≥3, 0%; per Lee scale). To treat CRS, 15% of pts required tocilizumab and 3% required steroids. Any grade serious neurologic events (NEs; per CTCAE v4.03) occurred in 10% of pts; 2% had Gr ≥3 and all recovered (1 pt developed Gr 4 immune-effector cell neurotoxicity syndrome related to tisa-cel concomitant with possible HHV6 encephalitis; 1 pt developed Gr 3 delirium unrelated to tisa-cel after progression of disease and start of salvage therapy). Median time to CRS and serious NEs onset were 4 and 8.5 d, respectively, with respective median time to resolution of 4 and 2 d. Three pts died from progressive disease; no deaths were treatment-related. Conclusions: Preliminary data from ELARA suggest that tisa-cel is effective in extensively treated r/r FL, resulting in high CRRs and ORRs, including in high-risk pts. The overall safety profile is favorable, with no severe CRS and very low NE reported, requiring limited anticytokine therapy. Updated safety and efficacy results on 97 pts (including dose and cellular kinetics data) will be presented at the meeting. Disclosures Fowler: TG Therapeutics: Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding; Roche: Research Funding. Dickinson:Roche: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Gilead: Consultancy, Honoraria, Research Funding, Speakers Bureau; Merck Sharp & Dohme: Consultancy; Novartis: Consultancy, Honoraria, Research Funding, Speakers Bureau; Janssen: Consultancy, Honoraria, Speakers Bureau. Dreyling:Astra Zeneca: Consultancy; Abbvie: Research Funding; Roche: Consultancy, Research Funding, Speakers Bureau; Celgene: Consultancy, Research Funding, Speakers Bureau; Bayer: Consultancy, Speakers Bureau; Beigene: Consultancy; Gilead: Consultancy, Research Funding, Speakers Bureau; Janssen: Consultancy, Research Funding, Speakers Bureau; Novartis: Consultancy. Martinez-Lopez:Janssen: Consultancy, Honoraria; Novartis: Consultancy; Janssen-cilag: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; BMS: Consultancy, Research Funding; Incyte: Consultancy, Research Funding. Kolstad:Merck: Research Funding; Nordic Nanovector: Membership on an entity's Board of Directors or advisory committees, Research Funding. Popplewell:Pfizer: Research Funding; Novartis: Research Funding; Roche: Research Funding. Chavez:Genentech: Speakers Bureau; Epizyme: Speakers Bureau; Gilead: Consultancy; Verastem: Consultancy; AbbVie: Consultancy; Karyopharm: Consultancy; BeiGene: Speakers Bureau; Novartis: Consultancy; Kite, a Gilead Company: Consultancy, Speakers Bureau; Pfizer: Consultancy; AstraZeneca: Speakers Bureau; Morphosys: Consultancy, Speakers Bureau; Merck: Research Funding; Bayer: Consultancy; Celgene: Consultancy. Bachy:Beigene: Membership on an entity's Board of Directors or advisory committees; Roche, Celgene, Amgen, Janssen, Gilead, Novartis, Sanofi: Honoraria; Amgen: Research Funding; Roche, Gilead: Consultancy. Kato:AbbVie, AstraZeneca, Celgene, Chugai, Eisai, Janssen, and Novartis: Consultancy; hugai, Takeda, Kyowa-Kirin, Abbvie, Novartis, Eisai, Janssen, Celgene, Ono: Research Funding; Chugai, Takeda, MSD, Kyowa-Kirin, Janssen, Celgene, Ono, Mundi, Dainippon-Sumitomo, Novartis: Honoraria. Harigae:Novartis, Chugai, BMS: Honoraria. Kersten:BMS: Consultancy, Honoraria, Other: TRAVEL, ACCOMMODATIONS, EXPENSES (paid by any for-profit health care company); Celgene: Consultancy, Honoraria, Other: TRAVEL, ACCOMMODATIONS, EXPENSES (paid by any for-profit health care company), Research Funding; Amgen: Consultancy, Honoraria, Other: TRAVEL, ACCOMMODATIONS, EXPENSES (paid by any for-profit health care company); Janssen/Cilag: Consultancy, Honoraria, Other: TRAVEL, ACCOMMODATIONS, EXPENSES (paid by any for-profit health care company); Miltenyi Biotech: Consultancy, Honoraria, Other: TRAVEL, ACCOMMODATIONS, EXPENSES (paid by any for-profit health care company); Takeda: Research Funding; Roche: Consultancy, Honoraria, Other: TRAVEL, ACCOMMODATIONS, EXPENSES (paid by any for-profit health care company), Research Funding; Kite/Gilead: Consultancy, Honoraria, Other: TRAVEL, ACCOMMODATIONS, EXPENSES (paid by any for-profit health care company); MSD: Consultancy, Honoraria, Other: TRAVEL, ACCOMMODATIONS, EXPENSES (paid by any for-profit health care company); Novartis: Consultancy, Honoraria, Other: TRAVEL, ACCOMMODATIONS, EXPENSES (paid by any for-profit health care company). Andreadis:Novartis: Research Funding; Gilead/Kite: Consultancy; Merck: Research Funding; Incyte: Consultancy; Karyopharm: Honoraria; Jazz Pharmaceuticals: Honoraria; BMS/Celgene/Juno: Honoraria, Research Funding; Genentech: Consultancy, Current equity holder in publicly-traded company. Riedell:Bayer: Honoraria; Karyopharm Therapeutics: Honoraria; Morphosys: Research Funding; Celgene/Bristol-Myers Squibb Company: Honoraria, Research Funding; Verastem Oncology: Honoraria; Kite Pharmaceuticals/Gilead: Honoraria, Research Funding; Novartis: Consultancy, Honoraria, Research Funding. Nastoupil:Karus Therapeutics: Research Funding; Gamida Cell: Honoraria; Gilead/KITE: Honoraria; Novartis: Honoraria, Research Funding; Merck: Research Funding; Celgene: Honoraria, Research Funding; Pfizer: Honoraria, Research Funding; Bayer: Honoraria; Janssen: Honoraria, Research Funding; LAM Therapeutics: Research Funding; TG Therapeutics: Honoraria, Research Funding; Genentech, Inc.: Honoraria, Research Funding. von Tresckow:Takeda: Honoraria, Other: Travel support, Research Funding; Amgen: Honoraria; Pfizer: Honoraria; Kite/Gilead: Honoraria; Roche: Honoraria; MSD Sharp & Dohme: Honoraria, Research Funding; Takeda: Honoraria, Other: Travel support, Research Funding; Novartis: Other: Travel support, Research Funding. Ferreri:Morphosys: Research Funding; Hutchinson: Research Funding; BMS: Research Funding; Gilead: Membership on an entity's Board of Directors or advisory committees, Research Funding. Teshima:Sharp & Dohme Corp: Consultancy, Honoraria; Bristol-Myers Squibb: Honoraria; Pfizer Japan Inc.: Honoraria; Kyowa Kirin Co., Ltd.: Honoraria, Research Funding; TEIJIN PHARMA LIMITED: Honoraria; The Center of Innovation Program from Japan Science and Technology Agency: Other; Fuji Pharma Co., Ltd.: Honoraria; NIPPON SHINYAKU CO., LTD.: Honoraria; Janssen Pharmaceutical K.K.: Other; Japan Society for the Promotion of Science KAKENHI (17H04206): Other; Novartis Pharma K.K.: Consultancy, Other: Manuscript preparation, Research Funding; Takeda Pharmaceutical Company Limited: Consultancy, Honoraria; Chugai Pharmaceutical Co., Ltd.: Research Funding; Astellas Pharma Inc.: Research Funding; Merck: Consultancy, Honoraria; Sanofi K.K.: Research Funding. Patten:Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Roche: Consultancy, Honoraria; AbbVie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Astra Zeneca: Honoraria; Gilead: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Consultancy, Honoraria. McGuirk:Novartis: Research Funding; Fresenius Biotech: Research Funding; Astellas: Research Funding; Juno Therapeutics: Consultancy, Honoraria, Research Funding; Allo Vir: Consultancy, Honoraria, Research Funding; Kite Pharmaceuticals: Consultancy, Honoraria, Research Funding, Speakers Bureau; Pluristem Ltd: Research Funding; Gamida Cell: Research Funding; Bellicum Pharmaceutical: Research Funding. Petzer:Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees; Kite-Gilead: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees; Roche: Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees. Viardot:Amgen: Honoraria, Other: advisory board; Novartis: Honoraria, Other: advisory board; Kite/Gilead: Honoraria, Other: advisory board; Roche: Honoraria, Other: advisory board. Zinzani:ADC Therapeutics: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; MSD: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Immune Design: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; BMS: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Merck: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; EUSA Pharma: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Immune Design: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Roche: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Incyte: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Sanofi: Consultancy, Membership on an entity's Board of Directors or advisory committees; Sandoz: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Portola: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Kyowa Kirin: Consultancy, Speakers Bureau; Eusapharma: Consultancy, Speakers Bureau; Verastem: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Kirin Kyowa: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Servier: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Gilead: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; TG Therapeutics, Inc.: Honoraria, Speakers Bureau; AbbVie: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Janssen-Cilag: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Janssen: Consultancy, Honoraria, Speakers Bureau; Celltrion: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Malladi:Novartis: Consultancy, Honoraria. Bubuteishvili Pacaud:Novartis: Current Employment. Forcina:Novartis: Current Employment. Zia:Novartis: Current Employment. Schuster:Novartis, Genentech, Inc./ F. Hoffmann-La Roche: Research Funding; AlloGene, AstraZeneca, BeiGene, Genentech, Inc./ F. Hoffmann-La Roche, Juno/Celgene, Loxo Oncology, Nordic Nanovector, Novartis, Tessa Therapeutics: Consultancy, Honoraria. Thieblemont:Cellectis: Speakers Bureau; Roche, Amgen, Kyte Gilead, Celgene, Abbvie, Novartis, Cellectis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel support; Roche, Hospita: Research Funding.
- Published
- 2020
20. The architecture of neoplastic follicles in follicular lymphoma; analysis of the relationship between the tumor and follicular helper T cells
- Author
-
Robert Marcus, Marta Pasikowska, Elizabeth H Phillips, William Townsend, Andrea Pepper, Deborah Yallop, Stephen Devereux, Piers E.M. Patten, and Jonathan R. Salisbury
- Subjects
endocrine system ,T Follicular Helper Cells ,Non-Hodgkin Lymphoma ,Follicular lymphoma ,law.invention ,Pathogenesis ,Confocal microscopy ,law ,Follicular phase ,Activation-induced (cytidine) deaminase ,medicine ,Humans ,Lymphoma, Follicular ,B-Lymphocytes ,Manchester Cancer Research Centre ,biology ,Immunological synapse formation ,ResearchInstitutes_Networks_Beacons/mcrc ,Germinal center ,Articles ,T-Lymphocytes, Helper-Inducer ,Hematology ,Germinal Center ,medicine.disease ,biology.protein ,Cancer research ,Lymph - Abstract
CD4+ T-follicular helper cells are essential for the survival, proliferation, and differentiation of germinal center B-cells and have been implicated in the pathogenesis of follicular lymphoma. To further define the role of these cells in follicular lymphoma, we used multiparameter confocal microscopy to compare the architecture of normal and neoplastic follicles and next generation sequencing to analyze the T-cell receptor repertoire in follicular lymphoma lymph nodes. Multiparameter analysis of lymph nodes showed that the proportion of T-follicular helper cells in normal and neoplastic follicles is the same and that the previously reported increase in T-follicular helper cell numbers in follicular lymphoma is thus due to an increase in the number and not content of follicles. As in normal germinal centers, T-follicular helper cells were shown to have a close spatial correlation with proliferating B-cells in neoplastic follicles, where features of immunological synapse formation were observed. The number of T-follicular helper cells in follicular lymphoma correlate with the rate of B-cell proliferation and T-follicular helper cells co-localized to Activation Induced Cytidine Deaminase expressing proliferating B-cells. T-cell receptor repertoire analysis of follicular lymphoma lymph nodes revealed that follicular areas are significantly more clonal when compared to the rest of the lymph node. These novel findings show that neoplastic follicles and germinal centers share important structural features and provide further evidence that T-follicular helper cells may play a role in driving B-cell proliferation and genomic evolution in follicular lymphoma. Our results also suggest that targeting this interaction would be an attractive therapeutic option.
- Published
- 2019
21. Interim results of the safety and immune-efficacy of 1 versus 2 doses of COVID-19 vaccine BNT162b2 for cancer patients in the context of the UK vaccine priority guidelines
- Author
-
Sophie Papa, Thomas Hayday, Carl Graham, Rosalind Graham, Miguel Muñoz-Ruiz, Muhammad Shamim Khan, Maria Conde Poole, Irene del Molino del Barrio, Elizabeth Harvey-Jones, Leticia Monin-Aldama, Francesca Di Rosa, Jeffrey Seow, Puay Lee, Shubhankar Sinha, Katie J. Doores, Helen Kakkassery, Sultan Abdul-Jawad, Anne Rigg, Shraddha Kamdar, Clara Domingo Vila, Sheeba Irshad, Thanussuyah Alaguthurai, Paul Fields, Timothy Tree, Ana Montes, James Spicer, Richard Davis, Jack Cooper, Isaac Francos Quijorna, Liane Dupont, Jennifer Vidler, Michael H. Malim, Piers E.M. Patten, Yin Wu, Magdalene Joseph, Mark Harries, Daniel Davies, Adrian Hayday, Duncan R. McKenzie, Josephine Eum, and Adam Laing
- Subjects
education.field_of_study ,medicine.medical_specialty ,business.industry ,Population ,Cancer ,Context (language use) ,Immune dysregulation ,medicine.disease_cause ,medicine.disease ,Vaccine efficacy ,Herd immunity ,Vaccination ,Internal medicine ,Medicine ,Seroconversion ,business ,education - Abstract
BackgroundThe efficacy and safety profile of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have not been definitively established in immunocompromised patient populations. Patients with a known cancer diagnosis were hitherto excluded from trials of the vaccines currently in clinical use.MethodsThis study presents data on the safety and immune efficacy of the BNT162b2 (Pfizer-BioNTech) vaccine in 54 healthy controls and 151 mostly elderly patients with solid and haematological malignancies, respectively, and compares results for patients who were boosted with BNT162b2 at 3 weeks versus those who were not. Immune efficacy was measured as antibody seroconversion, T cell responses, and neutralisation of SARS-CoV-2 Wuhan strain and of a variant of concern (VOC) (B.1.1.7). We also collected safety data for the BNT162b2 vaccine up to 5 weeks following first dose.FindingsThe vaccine was largely well tolerated. However, in contrast to its very high performance in healthy controls (>90% efficacious), immune efficacy of a single inoculum in solid cancer patients was strikingly low (below 40%) and very low in haematological cancer patients (below 15%). Of note, efficacy in solid cancer patients was greatly and rapidly increased by boosting at 21-days (95% within 2 weeks of boost). Too few haematological cancer patients were boosted for clear conclusions to be drawn.ConclusionsDelayed boosting potentially leaves most solid and haematological cancer patients wholly or partially unprotected, with implications for their own health; their environment and the evolution of VOC strains. Prompt boosting of solid cancer patients quickly overcomes the poor efficacy of the primary inoculum in solid cancer patients.RESEARCH IN CONTEXTEvidence before this studySome cancer patients have been shown to exhibit sustained immune dysregulation, inefficient seroconversion and prolonged viral shedding as a consequence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Consequently, their exclusion and, in particular, the exclusion of patients receiving systemic anti-cancer therapies, from the registry trials of the 5 approved COVID-19 vaccines raises questions about the efficacy and safety of SARS-CoV-2 vaccination in this patient population. In addition, whilst the change in the UK’s dosing interval to 12-weeks aimed to maximise population coverage, it is unclear whether this strategy is appropriate for cancer patients and those on systemic anti-cancer therapies.Added value of this studyWe report that the RNA-based SARS-CoV-2 BNT162b2 vaccine administered in cancer patients was well tolerated, and we provide first insights into both antibody and T cell responses to the vaccine in an immunocompromised patient population.Implications of all the available evidenceIn cancer patients, one dose of 30ug of BNT162b2 yields poor vaccine efficacy, as measured by seroconversion rates, viral neutralisation capacity and T cell responses, at 3- and 5-weeks following the first inoculum. Patients with solid cancers exhibited a significantly greater response following a booster at 21-days. These data support prioritisation of cancer patients for an early (21-day) second dose of the BNT162b2 vaccine. Given the globally poor responses to vaccination in patients with haematological cancers, post-vaccination serological testing, creation of herd immunity around these patients using a strategy of ‘ring vaccination’, and careful follow-up should be prioritised.
- Published
- 2021
22. Acute Immune Signatures and Their Legacies in Severe Acute Respiratory Syndrome Coronavirus-2 Infected Cancer Patients
- Author
-
Sheeba Irshad, Paul Fields, Doraid Alrifai, Richard Davis, Yin Wu, Isaac Francos Quijorna, Anna Lorenc, Eva Bugallo-Blanco, Julie Nuo En Chan, Anne Rigg, Jennifer Vidler, Adrian Hayday, Aadil A. Khan, Julien de Naurois, Charlotte Moss, Sultan Abdul-Jawad, Pierre Vantourout, Louisa Mcdonald, Duncan R. McKenzie, Mieke Van Hemelrijck, Paul R. Barber, Irene del Molino del Barrio, Sophie Papa, Katharine Bailey, Deborah Enting, Beth Russell, Sophie Hazell, Sarah Gee, Tony Ng, Sarah Ryan, You Zhou, Anthony C. C. Coolen, Iva Zlatareva, Piers E.M. Patten, Adam Laing, Sophia N. Karagiannis, Miguel Muñoz-Ruiz, Luca Bau, Thomas Hayday, Yadanar Lwin, Shraddha Kamdar, Matthew Fish, Reuben Benjamin, Rozalyn Yorke, Aislinn Jennings, Katie J. Doores, Mayur Kumar, Thanussuyah Alaguthurai, Leticia Monin, Mark Rowley, and James Spicer
- Subjects
0301 basic medicine ,Male ,Cancer Research ,viruses ,T-Lymphocytes ,medicine.disease_cause ,Severe Acute Respiratory Syndrome ,0302 clinical medicine ,vaccine ,Nasopharynx ,Neoplasms ,Medicine ,antibodies ,Young adult ,Aged, 80 and over ,education.field_of_study ,hemato-oncological ,biology ,Middle Aged ,Oncology ,030220 oncology & carcinogenesis ,Hematologic Neoplasms ,Female ,Antibody ,Adult ,Population ,Article ,virus shedding ,Immunophenotyping ,03 medical and health sciences ,Young Adult ,Immune system ,cancer ,Humans ,Viral shedding ,Seroconversion ,education ,seroconversion ,Aged ,business.industry ,SARS-CoV-2 ,Cancer ,COVID-19 ,Cell Biology ,Immune dysregulation ,medicine.disease ,030104 developmental biology ,Immunology ,biology.protein ,immune ,business - Abstract
Given the immune system's importance for cancer surveillance and treatment, we have investigated how it may be affected by SARS-CoV-2 infection of cancer patients. Across some heterogeneity in tumor type, stage, and treatment, virus-exposed solid cancer patients display a dominant impact of SARS-CoV-2, apparent from the resemblance of their immune signatures to those for COVID-19+ non-cancer patients. This is not the case for hematological malignancies, with virus-exposed patients collectively displaying heterogeneous humoral responses, an exhausted T cell phenotype and a high prevalence of prolonged virus shedding. Furthermore, while recovered solid cancer patients' immunophenotypes resemble those of non-virus-exposed cancer patients, recovered hematological cancer patients display distinct, lingering immunological legacies. Thus, while solid cancer patients, including those with advanced disease, seem no more at risk of SARS-CoV-2-associated immune dysregulation than the general population, hematological cancer patients show complex immunological consequences of SARS-CoV-2 exposure that might usefully inform their care., Graphical Abstract, Following SARS-Cov-2 infection, Sultan et al. showed that the majority of solid cancer patients cleared virus, recovered from COVID-19, and re-established their prior immunological status. In contrast, hematological cancer patients demonstrated delayed or negligible seroconversion, prolonged shedding, and sustained immune dysregulation highlighting the need for careful oversight of these patients.
- Published
- 2021
- Full Text
- View/download PDF
23. Humoral and Cellular Immunity to SARS-CoV-2 Vaccination in Cancer Patients: Completed Prospective Observational Study Using BNT162b2 mRNA Vaccine
- Author
-
Sultan Abdul-Jawad, Jennifer Vidler, Michael H. Malim, Carl Graham, Sophie Papa, Kamila Sychowska, Katie J. Doores, Sheeba Irshad, Thanussuyah Alaguthurai, Rosalind Graham, Charalampos Gousis, Jack Cooper, Paul Fields, Thomas Lechmere, Piers E.M. Patten, Bernard V. North, Adrian Hayday, Timothy Tree, Duncan R. McKenzie, Clara Domingo-Vila, Catherine Tremain, Jeffrey Seow, Angela Swampillai, Emily Pollock, Kasia Owzarczyk, Leticia Monin-Aldama, Miguel Muñoz-Ruiz, and Hartmut Kristeleit
- Subjects
Chemotherapy ,Cellular immunity ,medicine.medical_specialty ,business.industry ,Immunogenicity ,medicine.medical_treatment ,Cancer ,Context (language use) ,medicine.disease ,Vaccination ,Internal medicine ,Clinical endpoint ,Medicine ,Seroconversion ,business - Abstract
Background: Cancer patients are vulnerable populations for COVID-19 complications and mortality. We previously reported on the poor single-dose immunogenicity of BNT162b2 mRNA vaccine in cancer patients, particularly those with haematological malignancies. Methods: In this prospective, observational study relating to the safety and immunogenicity of BNT162b2 mRNA vaccine, 201 vaccinated cancer patients (solid cancer n=125; haematological cancer n=76) and 54 healthy controls (mostly health-care workers “HCW”) were recruited between December 8th, 2020, and April 23rd, 2021. The previously reported interim results covered a period of 101 days since first patient recruitment, during which time 47 subjects received a second “boost” vaccination on day 21. Because of the change in UK Government policy, all others received a delayed vaccine boost at about 12 weeks after their first vaccination, and had their blood sampled 2 weeks’ later. Here, we describe immunogenicity data following the delayed boost in 31 HCWs, 72 solid cancer and 56 haematological cancer patients. Seroconversion, virus neutralisation, and T cell assays were as described previously, with an additional test for neutralisation of the B.1.617.2 (delta) variant-ofconcern (VOC). The primary endpoint of the study was the impact on seroconversion following delayed (>21days) vaccine boosting in solid and haematological cancer patients. The secondary endpoints were: safety following delayed vaccine boost; T cell responses; and neutralisation of SARS-CoV-2 Wuhan (“wild type” [WT]), B.1.1.7 (alpha), and B.1.617.2 (delta) variants. Findings: Delayed (>21days) boost vaccination of solid cancer patients and haematological cancer patients with the BNT162b2 vaccine was well tolerated, as the primary vaccination had been. There was no vaccine-associated death. Boosting significantly increased solid cancer patients’ seroconversion responses, that had been strikingly poor in response to a single dose: from 38% to 84%. Boosting also significantly improved vaccine immunogenicity for haematological cancer patients, but most (57%) still failed to seroconvert. Seroconversion correlated strongly with the capacity to neutralise SARSCoV- 2 cell entry, although neutralisation of the WT variant was typically greater than of the VOC. Neutralisation was significantly increased by boosting for HCWs but not for cancer patients. In comparison to seroconversion, boosting achieved higher rates of functional T cell responsiveness (de novo responses) but had little impact on the magnitude of T cell responses for those who had responded to first-dose vaccination. When patients were scored as showing both seroconversion and T cell responses, the unfavourable situation of haematological cancer patients was overt with only 36% (12/33) defined as being responders compared to 78% (25/32) of solid cancer patients and 88% (15/17) of HCWs. There was no significant difference in any aspect of immunogenicity for HCWs or solid cancer patients receiving the delayed boost versus the day 21 boost (this comparison could not be made for haematological cancer patients because too few received an early boost). Chemotherapy within 15 days either side of the boost exacerbated the likelihood of non-responsiveness to the vaccine. Interpretation: Boosting at either 3 weeks or longer (up to 12 weeks) post-primary vaccination shows high efficacy in terms of seroconversion of solid cancer patients and increases in their SARS-CoV-2 Spike-specific antibody titres. By contrast, delayed boosting left most haematological cancer patients without serological protection against SARS-CoV-2 infection. These data support the ongoing adjustment of health care measures to limit the evident vulnerability of such individuals to SARS-CoV- 2, and to limit their potential to transmit virus variants that might develop in the context of absent or partial immunoprotection. The absence of any clear improvements in immunogenicity of a delayed boost relative to boosting on day 21 emphasizes the importance of early boosting for cancer patients, and potentially of doing so repeatedly, particularly given how well the vaccine was tolerated. Chemotherapy, if possible should be withheld 15 days before and 15 days after the vaccination date. Trial Registration: The trial is registered with the NHS Health Research Authority (HRA) and Health and Care Research Wales (HCRW) (REC ID: 20/HRA/2031). Funding: KCL, CRUK, Leukemia & Lymphoma Society, Wellcome Trust, Rosetrees Trust, Francis Crick Institute. Declaration of Interest: None to declare. Ethical Approval: The trial was approved by the institutional review boards of the participating institutions (IRAS ID: 282337 REC ID: 20/HRA/2031).
- Published
- 2021
24. Gene-edited healthy donor CAR T cells show superior anti-tumour activity compared to CAR T cells derived from patients with lymphoma in an in vivo model of high-grade lymphoma
- Author
-
Piers E.M. Patten, Ana M Metelo, Carlo Scala, Glenda Dickson, Ruby Quartey-Papafio, Maria Almena-Carrasco, Reuben Benjamin, Elisa Peranzoni, Farzin Farzaneh, Orla Stewart, Alan G. Ramsay, Thomas Pertel, Charlotte Graham, Nikolaos Ioannou, Agnieszka Jozwik, Sandra Dupouy, and Andrea Pepper
- Subjects
Cancer Research ,High-grade lymphoma ,Lymphoma, B-Cell ,Letter ,Receptors, Antigen, T-Cell, alpha-beta ,Antigens, CD19 ,Immunotherapy, Adoptive ,Anti tumour ,Mice ,Text mining ,In vivo ,Medicine ,Animals ,Humans ,Gene ,Cells, Cultured ,Gene Editing ,business.industry ,Hematology ,medicine.disease ,Healthy Volunteers ,Lymphoma ,Disease Models, Animal ,Oncology ,Cancer research ,Tumour immunology ,Immunotherapy ,Healthy donor ,Car t cells ,business - Published
- 2020
25. Poor outcome and prolonged persistence of SARS‐CoV‐2 RNA in COVID‐19 patients with haematological malignancies; King's College Hospital experience
- Author
-
Judith C. W. Marsh, Piers E.M. Patten, Daniele Avenoso, Reuben Benjamin, James Galloway, Carmel Rice, Sam Norton, Victoria Potter, Ghulam J. Mufti, Antonio Pagliuca, Vallari Shah, Jennifer Vidler, Anita Sarma, Shreyans Gandhi, Deborah Yallop, Mark Zuckerman, Thinzar Ko Ko, M. Mansour Ceesay, Varun Mehra, R. Sanderson, Andrea Kuhnl, Austin G. Kulasekararaj, Hugues de Lavallade, Sobia Sharif, and Pramila Krishnamurthy
- Subjects
medicine.medical_specialty ,Letter ,Coronavirus disease 2019 (COVID-19) ,Adverse outcomes ,business.industry ,viruses ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Hematology ,Hospital experience ,Persistence (computer science) ,03 medical and health sciences ,0302 clinical medicine ,Increased risk ,030220 oncology & carcinogenesis ,Internal medicine ,Medicine ,Letters ,business ,Survival rate ,030215 immunology ,Cohort study - Abstract
Severe acute respiratory syndrome coronavirus‐2 (SARS‐CoV‐2), emerged at the end of 2019 and caused an infection named COVID‐19 (Guan, Ni et al. 2020). Patients with compromised immune systems are at increased risk of complications but this risk is not precisely defined (Liang, Guan et al. 2020). Although age, gender, comorbidities and ethnicity are risk factors for adverse outcomes (Huang, Wang et al. 2020), various pre‐existing conditions, including haematological cancers, have also been reported to correlate with poor outcomes (Aries, Davies et al. 2020, He, Chen et al. 2020, Malard, Genthon et al. 2020, Martin‐Moro, Marquet et al. 2020, medRxiv 2020)
- Published
- 2020
26. Outcomes of COVID-19 in patients with CLL: a multicenter international experience
- Author
-
Neil Bailey, Fatima Miras, Jose Angel Hernandez-Rivas, Chadi Nabhan, John M. Pagel, Elise A. Chong, Manali Kamdar, Sigrid S. Skånland, Raul Cordoba, Matthew S. Davids, Mazyar Shadman, Angus Broom, Ellin Berman, Shuo Ma, Anthony R. Mato, Paul M. Barr, Meera Patel, Lindsey E. Roeker, Erlene K. Seymour, José A. García-Marco, Andrew D. Zelenetz, Anders Österborg, Matthew R. Wilson, Toby A. Eyre, Danielle M. Brander, Krista Isaac, Jeffrey Pu, Mark B. Geyer, Richard R. Furman, Sonia Lebowitz, Renata Walewska, Talha Munir, Nikita Malakhov, John N. Allan, Scott F. Huntington, Inhye E. Ahn, Darko Antic, Lotta Hanson, Adrian Wiestner, Ryan Jacobs, Paola Ghione, Nicolas Martinez-Calle, Lori A. Leslie, Erica Bhavsar, Suchitra Sundaram, Daniel Wojenski, Jennifer R. Brown, Chaitra S. Ujjani, Amanda N. Seddon, Daniel Naya, Javier López-Jiménez, Harriet S. Walter, Christine E. Ryan, Craig A. Portell, Krish Patel, Dima El-Sharkawi, Michael Koropsak, Guilherme Fleury Perini, Noemi Fernandez Escalada, Helen Parry, Nicole Lamanna, and Piers E.M. Patten
- Subjects
0301 basic medicine ,Male ,Clinical Trials and Observations ,Chronic lymphocytic leukemia ,Anti-Inflammatory Agents ,Disease ,Biochemistry ,law.invention ,0302 clinical medicine ,law ,Case fatality rate ,Agammaglobulinaemia Tyrosine Kinase ,Aged, 80 and over ,Risk of infection ,Hematology ,Middle Aged ,Intensive care unit ,3. Good health ,Treatment Outcome ,030220 oncology & carcinogenesis ,Female ,Coronavirus Infections ,BLOOD Commentary ,Adult ,medicine.medical_specialty ,Immunology ,Pneumonia, Viral ,Antiviral Agents ,03 medical and health sciences ,Betacoronavirus ,Internal medicine ,medicine ,Humans ,Pandemics ,Protein Kinase Inhibitors ,Survival analysis ,COVID-19 Serotherapy ,Aged ,business.industry ,SARS-CoV-2 ,Immunization, Passive ,COVID-19 ,Cell Biology ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Survival Analysis ,Clinical trial ,Pneumonia ,030104 developmental biology ,business - Abstract
There is a Blood Commentary on this article in this issue., Key Points Both watch-and-wait and treated CLL patients have high mortality rates when admitted for COVID-19. Receiving a BTKi for CLL at COVID-19 diagnosis severe enough to require hospitalization did not influence case fatality rate in this study., Given advanced age, comorbidities, and immune dysfunction, chronic lymphocytic leukemia (CLL) patients may be at particularly high risk of infection and poor outcomes related to coronavirus disease 2019 (COVID-19). Robust analysis of outcomes for CLL patients, particularly examining effects of baseline characteristics and CLL-directed therapy, is critical to optimally manage CLL patients through this evolving pandemic. CLL patients diagnosed with symptomatic COVID-19 across 43 international centers (n = 198) were included. Hospital admission occurred in 90%. Median age at COVID-19 diagnosis was 70.5 years. Median Cumulative Illness Rating Scale score was 8 (range, 4-32). Thirty-nine percent were treatment naive (“watch and wait”), while 61% had received ≥1 CLL-directed therapy (median, 2; range, 1-8). Ninety patients (45%) were receiving active CLL therapy at COVID-19 diagnosis, most commonly Bruton tyrosine kinase inhibitors (BTKi’s; n = 68/90 [76%]). At a median follow-up of 16 days, the overall case fatality rate was 33%, though 25% remain admitted. Watch-and-wait and treated cohorts had similar rates of admission (89% vs 90%), intensive care unit admission (35% vs 36%), intubation (33% vs 25%), and mortality (37% vs 32%). CLL-directed treatment with BTKi’s at COVID-19 diagnosis did not impact survival (case fatality rate, 34% vs 35%), though the BTKi was held during the COVID-19 course for most patients. These data suggest that the subgroup of CLL patients admitted with COVID-19, regardless of disease phase or treatment status, are at high risk of death. Future epidemiologic studies are needed to assess severe acute respiratory syndrome coronavirus 2 infection risk, these data should be validated independently, and randomized studies of BTKi’s in COVID-19 are needed to provide definitive evidence of benefit., Visual Abstract
- Published
- 2020
27. In vitro and in vivo evidence for uncoupling of B-cell receptor internalization and signaling in chronic lymphocytic leukemia
- Author
-
Elizabeth H Phillips, Piers E.M. Patten, Kirsty Cuthill, Eve Coulter, Silvia Mele, William Townsend, Andrea Pepper, Najeem'deen Folarin, and Stephen Devereux
- Subjects
0301 basic medicine ,Manchester Cancer Research Centre ,Chemistry ,Endosome ,ResearchInstitutes_Networks_Beacons/mcrc ,Chronic lymphocytic leukemia ,media_common.quotation_subject ,Hematology ,medicine.disease ,Article ,Cell biology ,03 medical and health sciences ,Leukemia ,030104 developmental biology ,Journal Article ,medicine ,Chronic Lymphocytic Leukemia ,Signal transduction ,Internalization ,Receptor ,B cell receptor internalization ,Tyrosine kinase ,media_common - Abstract
B-cell receptor activation, occurring within lymph nodes, plays a key role in the pathogenesis of chronic lymphocytic leukemia and is linked to prognosis. As well as activation of downstream signaling, receptor ligation triggers internalization, transit to acidified endosomes and degradation of ligand-receptor complexes. Herein, we investigated the relationship between these two processes in normal and leukemic B cells. We found that leukemic B cells, particularly anergic cases lacking the capacity to initiate downstream signaling, internalize and accumulate ligand in acidified endosomes more efficiently than normal B cells. Furthermore, ligation of either surface CD79B, a B-cell receptor component required for downstream signaling, or surface Immunoglobulin M (IgM) by cognate agonistic antibody, showed that the two molecules internalize independently of each other in leukemic but not normal B cells. Since association with surface CD79B is required for surface retention of IgM, this suggests that uncoupling of B-cell receptor internalization from signaling may be due to the dissociation of these two molecules in leukemic cells. A comparison of lymph node with peripheral blood cells from chronic lymphocytic leukemia patients showed that, despite recent B-cell receptor activation, lymph node B cells expressed higher levels of surface IgM. This surprising finding suggests that the B-cell receptors of lymph node- and peripheral blood-derived leukemic cells might be functionally distinct. Finally, long-term therapy with the Bruton’s tyrosine kinase inhibitors ibrutinib or acalabrutinib resulted in a switch to an anergic pattern of B-cell receptor function with reduced signaling capacity, surface IgM expression and more efficient internalization.
- Published
- 2017
28. Expansion of T Follicular Helper Cells in NLC Co-Cultures Reinforces the Concept of Co-Evolution of CLL and Supportive T Helper Cell Clones
- Author
-
Qing Ma, William G. Wierda, Alan G. Ramsay, Karen Clise-Dwyer, Zeev Estrov, Alicia Vaca, Jan A. Burger, Mariela Sivina, Alessandra Ferrajoli, Piers E.M. Patten, Ekaterina Kim, Daniel Li, Elisavet Vlachonikola, and Nikolaos Ioannou
- Subjects
medicine.anatomical_structure ,Immunology ,Follicular phase ,medicine ,Cell Biology ,Hematology ,T helper cell ,Biology ,Biochemistry - Abstract
CLL cells proliferate in secondary lymphatic tissues where the CLL cells interact with T cells, monocyte-derived nurselike cells and mesenchymal stromal cells, collectively referred to as the tissue microenvironment. The CLL lymph node morphology with proliferation centers resembles normal B cell follicles, suggesting that mechanisms regulating the expansion of antigen-selected normal B cells during the germinal center (GC) reaction also regulate CLL cell expansion. Normal GC reactions require specialized T follicular helper (Tfh) cells, which provide T cell help to antigen-stimulated B cells. Another important T cell subset are Tregs which regulate immune tolerance and homeostasis. NLC co-culture have been used to dissect cellular and molecular interactions between CLL cells and the lymph node microenvironment in vitro. To gain insight into the role of T cell subsets in CLL, we utilized the NLC model to characterize T cell subsets and their dynamics in NLC co-cultures. We quantified CD4 + and CD8 + T cells in co-cultures from 18 different patients, observing that CD4 + T cell counts remained stable over time (449±62 at baseline to 443±80 cells/µl after 14 days). In contrast, numbers of CD8 + T cells declined from 180±42 cells/µl to 98±16 cells/µl on day 14 (p Disclosures Ferrajoli: Janssen: Other: Advisory Board ; AstraZeneca: Other: Advisory Board, Research Funding; BeiGene: Other: Advisory Board, Research Funding. Wierda: Oncternal Therapeutics, Inc.: Research Funding; Sunesis: Research Funding; Gilead Sciences: Research Funding; GSK/Novartis: Research Funding; Acerta Pharma Inc.: Research Funding; Genentech: Research Funding; Karyopharm: Research Funding; KITE Pharma: Research Funding; Juno Therapeutics: Research Funding; Pharmacyclics LLC, an AbbVie Company: Research Funding; Cyclacel: Research Funding; Miragen: Research Funding; Loxo Oncology, Inc.: Research Funding; Janssen: Research Funding; Xencor: Research Funding; AstraZeneca: Research Funding; Genzyme Corporation: Consultancy; AbbVie: Research Funding. Patten: ABBVIE: Honoraria; ASTRA ZENECA: Honoraria; GILEAD SCIENCES: Honoraria, Research Funding; JANSSEN: Honoraria; NOVARTIS: Honoraria; ROCHE: Research Funding. Burger: Novartis: Other: Travel/Accommodations/Expenses, Speakers Bureau; Beigene: Research Funding, Speakers Bureau; Pharmacyclics LLC: Consultancy, Other: Travel/Accommodations/Expenses, Research Funding, Speakers Bureau; Gilead: Consultancy, Other: Travel/Accommodations/Expenses, Research Funding, Speakers Bureau; TG Therapeutics: Other: Travel/Accommodations/Expenses, Research Funding, Speakers Bureau; AstraZeneca: Consultancy; Janssen: Consultancy, Other: Travel/Accommodations/Expenses, Speakers Bureau.
- Published
- 2021
29. Outcome for patients with relapsed/refractory aggressive lymphoma treated with gemcitabine and oxaliplatin with or without rituximab; a retrospective, multicentre study
- Author
-
Vijay Dhanapal, Chia Lianwea, Robert Marcus, David Wrench, Stephen Devereux, Stella Bowcock, Paul Fields, Deborah Yallop, Menaka Gunasekara, Shireen Kassam, Corinne De Lord, and Piers E.M. Patten
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Lymphoma ,Organoplatinum Compounds ,medicine.medical_treatment ,Aggressive lymphoma ,Deoxycytidine ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Refractory ,Recurrence ,Positron Emission Tomography Computed Tomography ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Aged ,Neoplasm Staging ,Retrospective Studies ,Aged, 80 and over ,Chemotherapy ,business.industry ,Hematology ,Middle Aged ,medicine.disease ,Gemcitabine ,Oxaliplatin ,Regimen ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Female ,Rituximab ,Tomography, X-Ray Computed ,business ,Febrile neutropenia ,030215 immunology ,medicine.drug - Abstract
The treatment of relapsed aggressive lymphoma remains a challenge. Platinum-containing chemotherapy is standard of care. Gemcitabine/oxaliplatin (Gem-Ox) with or without rituximab (R) is an outpatient regimen with a favorable toxicity profile. This retrospective 'real world' study reports outcomes for 44 unselected patients with relapsed/refractory aggressive lymphoma treated with Gem-Ox ± R. 41% had primary refractory disease. The overall response rate (ORR) was 43% with a complete response (CR) of 30%. Response to the prior treatment regimen significantly affected the ORR with only 8% achieving CR if prior remission was12 months. Grade 3-4 hematological toxicity was common and 22% had febrile neutropenia. Eight patients proceeded to stem cell transplant. Overall, outcomes remain poor with a median overall survival of 8 months. In this high-risk group of patients, Gem-Ox ± R results in similar responses to other more toxic, inpatient regimens and should therefore be considered as second line therapy in relapsed lymphoma.
- Published
- 2017
30. Colitis After CAR T-Cell Therapy for Refractory Large B-Cell Lymphoma Responds to Anti-Integrin Therapy
- Author
-
Andrea Kuhnl, Ahmed Hashim, Piers E.M. Patten, R. Sanderson, Marc L Ooft, and Bu'Hussain Hayee
- Subjects
Text mining ,Refractory ,business.industry ,Gastroenterology ,Cancer research ,medicine ,Immunology and Allergy ,CAR T-cell therapy ,Anti integrin ,Colitis ,business ,B-cell lymphoma ,medicine.disease - Published
- 2020
31. Continued Long Term Responses to Ibrutinib + Venetoclax Treatment for Relapsed/Refractory CLL in the Blood Cancer UK TAP Clarity Trial
- Author
-
Christopher Fegan, Francesco Forconi, Chhaya Sankhalpara, Stephen Devereux, Andrew R. Pettitt, Andy C. Rawstron, Nichola Webster, Francesca Yates, John G. Gribben, Peter Hillmen, Kristian Brock, Piers E.M. Patten, Alison McCaig, Christopher P. Fox, Rebecca H. Boucher, Surita Dalal, Anna Schuh, Adrian Bloor, and Donald Macdonald
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Venetoclax ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,Blood cancer ,chemistry.chemical_compound ,chemistry ,Internal medicine ,Ibrutinib ,Relapsed refractory ,Medicine ,business - Abstract
Background: The Blood Cancer UK TAP CLARITY trial for relapsed or refractory CLL combined IBR with VEN in order to eradicate detectable disease with a plan to stop therapy early if measurable residual disease (MRD) Aims: To provide long term follow up data for the combination of IBR and VEN for R/R CLL patients. IWCLL response status and MRD responses status at the 38 month time point are now presented in this abstract. Methods: CLARITY is a Phase II trial combining IBR with VEN in 50 patients with R/R CLL. After 2 months of IBR, VEN was added initially at a daily dose of 10mg or 20mg/day escalating weekly to a final daily dose of 400mg/day. CLL MRD was quantified using >6 colour ERIC-standard flow cytometry (detection limit 10-5/0.001%). Paired PB & BM samples were assessed at months 8, 14, 26 and longitudinal PB samples were taken at multiple time points. Fifty-four patients were recruited from May 2016 to November 2017. The median number of prior therapies was 1 (range: 1-6). 20% patients had del(17p); 25% del(11q); 75% had unmutated IGVH. Four patients that discontinued treatment due to ibrutinib-related adverse events in the first 8 weeks before starting VEN were replaced so that, in total, 50 patients received the combination of IBR and VEN. The interruption of therapy was indicated following confirmation of a CR according to the iwCLL2008 criteria and MRD reduction Results: Of the 50 evaluable patients recruited to the study, 23 patients stopped both treatments at or before M38; the majority due to achieving MRD4 (17/23). 27/50 patients were still receiving at least one trial treatment at this time point, with M38 response data pending for 11 patients. Of the 27 evaluable patients on treatment, the overall response rate (ORR), comprising of 10 patients with CR and 3 with CRi, was 81% compared to an ORR of 91% in the 36 patients receiving at least one trial treatment at M26 respectively. One additional patient discontinued treatment due to MRD negativity after M38. Of the 18 patients stopping due to MRD negativity, 14/18 continue to have Overall, MRD responses continued to improve after the first year of combination treatment with 24/50 (48%) patients achieving MRD4 in the BM at month 26 compared to 20/50 (40%) at month 14. After 4 months of combination, the median log CLL depletion was 2.9 (range 0.2-4.8) for patients achieving There was only one case of biochemical TLS. Other side-effects were mild and/or manageable, most commonly neutropenia (3/37 grade 2, 34/37 grade 3/4). Two Suspected Unexpected Serious Adverse Reactions (SUSARs) were reported (abdominal pain and pemphigus), 44 Serious Adverse Events (SAEs), and 1153 Adverse Events (AEs) (of which 141 were grade 3 or 4) were reported. Summary/Conclusion: After 38 months, the response to IBR+VEN is sustained despite planned discontinuation of therapy in MRD negative patients. The initial rate of disease depletion (during the first two months of combined IBR+VEN exposure) is highly predictive of longer-term response to combination IBR+VEN treatment in relapsed/refractory CLL. Patients who do not show rapid disease clearance and have persistent MRD after 12 months of combination IBR+VEN usually have stable or slowly decreasing disease levels akin to that seen in IBR monotherapy. These data will be updated at ASH 2020. Disclosures Hillmen: AbbVie: Speakers Bureau; Janssen: Speakers Bureau; Gilead: Speakers Bureau; Pharmacyclics: Other: Financial or Material spport; Morphosys: Other: Consulting fees; Sunesis: Other: Consulting fees. Brock:Invex: Other: Consulting and speaker fees; Merck: Other: Reimbursement of costs; Eli Lilly: Other: Consulting and speaker fees; AstraZeneca: Current equity holder in publicly-traded company; GlaxoSmithKline: Current equity holder in publicly-traded company. Schuh:Illumina: Other: Consulting fees; Abbvie: Other: Consulting fees; Gilead: Other: Consulting fees; Roche: Other: Consulting fees. Pettitt:Roche: Honoraria, Other: Hospitality, Research Funding; Napp: Research Funding; Celgene: Other: Hospitality, Research Funding; Chugai: Research Funding; Gilead: Honoraria, Other: Hospitality, Research Funding; Kite: Honoraria, Other: Hospitality, Research Funding; GSK: Research Funding; Novartis: Research Funding. Gribben:Janssen: Honoraria, Research Funding; AstraZeneca: Honoraria, Research Funding; Abbvie: Honoraria; Celgene: Research Funding. Patten:Roche: Consultancy, Honoraria; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen: Consultancy, Honoraria; Gilead: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Astra Zeneca: Honoraria; AbbVie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees. Bloor:Abbvie: Consultancy, Honoraria, Other: Conference Funding, Speakers Bureau; Janssen: Consultancy, Honoraria, Other: Conference Funding , Speakers Bureau. Fox:Celgene: Current equity holder in publicly-traded company; Sunesis: Current equity holder in publicly-traded company; Atarabio: Current equity holder in publicly-traded company; Abbvie: Current equity holder in private company, Current equity holder in publicly-traded company; Gilead: Current equity holder in private company, Current equity holder in publicly-traded company; Adienne: Current equity holder in private company, Current equity holder in publicly-traded company; Roche: Current equity holder in private company, Current equity holder in publicly-traded company; AstraZeneca: Current equity holder in publicly-traded company; Takeda: Current equity holder in private company, Current equity holder in publicly-traded company. Forconi:Roche: Honoraria; Janssen: Honoraria, Other: Fees for cosulting or advisory role, received travel and expenses, Speakers Bureau; AbbVie: Honoraria, Other: Fees for cosulting or advisory role, received travel and expenses, Speakers Bureau; Gilead: Research Funding; Astra Zeneca: Other: Fees for cosulting or advisory role; Menarini: Other: Fees for cosulting or advisory role; Novartis: Honoraria. Rawstron:BD Biosciences (Intrasure): Patents & Royalties. Hillmen:Alexion: Consultancy, Research Funding, Speakers Bureau; AstraZeneca: Consultancy, Speakers Bureau; Acerta: Other: Financial or material support; Roche: Consultancy, Other: Financial or material support, Research Funding, Speakers Bureau; AbbVie: Consultancy, Other: Financial or material support, Research Funding, Speakers Bureau; Pharmacyclics: Other: Financial or material support, Research Funding; Janssen: Consultancy, Other: Financial or material support, Research Funding, Speakers Bureau; Apellis: Consultancy, Research Funding, Speakers Bureau; Gilead: Other: Financial or material support, Research Funding.
- Published
- 2020
32. Ibrutinib Plus Venetoclax in Relapsed/Refractory Chronic Lymphocytic Leukemia: The CLARITY Study
- Author
-
Piers E.M. Patten, Rebecca Bishop, Andrew R. Pettitt, Christopher Fegan, Adrian Bloor, Samuel Muñoz-Vicente, Peter Hillmen, Kristian Brock, Christopher P. Fox, Rebecca H. Boucher, Andy C. Rawstron, Francesca Yates, Anna Schuh, Alison McCaig, Francesco Forconi, John G. Gribben, Talha Munir, Donald Macdonald, and Stephen Devereux
- Subjects
Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Chronic lymphocytic leukemia ,chemistry.chemical_compound ,Piperidines ,Recurrence ,Internal medicine ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Hematologic Malignancy ,Humans ,Aged ,Aged, 80 and over ,Sulfonamides ,Errata ,Venetoclax ,business.industry ,Adenine ,Middle Aged ,medicine.disease ,Bridged Bicyclo Compounds, Heterocyclic ,Leukemia, Lymphocytic, Chronic, B-Cell ,Pyrimidines ,chemistry ,Ibrutinib ,Relapsed refractory ,Pyrazoles ,Female ,business ,RAPID COMMUNICATION - Abstract
PURPOSE The treatment of chronic lymphocytic leukemia (CLL) has been revolutionized by targeted therapies that either inhibit proliferation (ibrutinib) or reactivate apoptosis (venetoclax). Both significantly improve survival in CLL and replace chemoimmunotherapy for many patients. However, individually, they rarely lead to eradication of measurable residual disease (MRD) and usually are taken indefinitely or until progression. We present the CLARITY trial that combined ibrutinib with venetoclax to eradicate detectable CLL with the intention of stopping therapy. PATIENTS AND METHODS CLARITY is a phase II trial that combined ibrutinib with venetoclax in patients with relapsed or refractory CLL. The primary end point was eradication of MRD after 12 months of combined therapy. Key secondary end points were response by International Workshop on CLL criteria, safety, and progression-free and overall survival. RESULTS In 53 patients after 12 months of ibrutinib plus venetoclax, MRD negativity (fewer than one CLL cell in 10,000 leukocytes) was achieved in the blood of 28 (53%) and the marrow of 19 (36%). Forty-seven patients (89%) responded, and 27 (51%) achieved a complete remission. After a median follow-up of 21.1 months, one patient progressed, and all patients were alive. A single case of biochemical tumor lysis syndrome was observed. Other adverse effects were mild and/or manageable and most commonly were neutropenia or GI events. CONCLUSION The combination of ibrutinib plus venetoclax was well tolerated in patients with relapsed or refractory CLL. There was a high rate of MRD eradication that led to the cessation of therapy in some patients. The progression-free and overall survival rates are encouraging for relapsed and refractory CLL.
- Published
- 2019
33. A retrospective analysis of post-transplant lymphoproliferative disorder following liver transplantation
- Author
-
Robert Marcus, P. Tachtatzis, Stephen Devereux, Shireen Kassam, John O'Grady, Piers E.M. Patten, Feras Al Fararjeh, Shameem Mahmood, Abid Suddle, Deborah Yallop, Nigel Heaton, and Kosh Agrawal
- Subjects
0301 basic medicine ,Adult ,Male ,medicine.medical_specialty ,Adolescent ,Databases, Factual ,medicine.medical_treatment ,Kaplan-Meier Estimate ,Liver transplantation ,Gastroenterology ,Post-transplant lymphoproliferative disorder ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,EBV Positive ,Retrospective analysis ,Humans ,Aged ,Neoplasm Staging ,Retrospective Studies ,Chemotherapy ,Hematology ,business.industry ,General Medicine ,Middle Aged ,medicine.disease ,Combined Modality Therapy ,Survival Analysis ,Lymphoproliferative Disorders ,Liver Transplantation ,030104 developmental biology ,Treatment Outcome ,Median time ,030220 oncology & carcinogenesis ,Rituximab ,Female ,business ,medicine.drug - Abstract
OBJECTIVE: To evaluate response rates and survival in adults developing post-transplant lymphoproliferative disorder (PTLD) following liver transplantation METHODS: Patients were identified retrospectively and data collected through local liver and haematology electronic databases and pharmacy records. RESULTS: Forty-five patients were identified. The median age at first transplant and at development of PTLD was 48 and 54 years respectively, with the median time from transplant to PTLD diagnosis of 56 months. The majority of cases (76%) were monomorphic B-cell lymphomas and 36% of tumours were EBV positive. Treatment involved reduction in immune-suppression (RIS) in 30 (67%) with RIS the only treatment in 3. Ten (22%) patients were treated with rituximab alone, 13 (29%) with chemotherapy alone and 14 (31%) patients were treated with rituximab and chemotherapy. Twenty-six (58%) patients achieved a complete response (CR). At a median follow-up of 27 months the median overall survival (OS) was 50 months Response and OS were not associated with clinical factors or the use of rituximab. CONCLUSION: Outcomes reported in this study are favourable and comparable to those reported previously. The addition of rituximab did not appear to have improved outcomes in this series, although a significant proportion of patients were able to avoid chemotherapy.
- Published
- 2017
34. Allogeneic Anti-CD19 CAR T Cells Manufactured from Healthy Donors Provide a Unique Cellular Product with Distinct Phenotypic Characteristics Compared to CAR T Cells Generated from Patients with Mature B Cell Malignancies
- Author
-
Andrea Pepper, Agnieszka Jozwik, Nikolaos Ioannou, Charlotte Graham, Orla Stewart, Ruby Quartey-Papafio, Sandra Dupouy, Reuben Benjamin, Farzin Farzaneh, Elisa Peranzoni, Alan G. Ramsay, Carlo Scala, Ana M Metelo, Glenda Dickson, Maria Almena-Carrasco, and Piers E.M. Patten
- Subjects
education.field_of_study ,biology ,business.industry ,CD3 ,T cell ,Immunology ,Population ,CD28 ,Cell Biology ,Hematology ,Biochemistry ,medicine.anatomical_structure ,Antigen ,Aldesleukin ,biology.protein ,medicine ,Cancer research ,IL-2 receptor ,education ,business ,CD8 - Abstract
Despite the success of autologous anti-CD19 CAR T cell therapy in B-Acute lymphoblastic leukaemia (B-ALL) and Diffuse Large B Cell Lymphoma (DLBCL), treatment failures occur. One contributing factor may be the intrinsic T cell fitness of the CAR T cell product that is influenced by the underlying malignancy and prior treatments. With the advent of gene editing, 'off the shelf' non-HLA matched healthy donor (HD) CAR T cells are under investigation for the treatment of patients (pts) in clinical trials. UCART19 (S68587) is a first-in-class allogeneic CAR T cell product expressing a second generation anti-CD19 CAR with TALEN®-mediated gene knockouts of T cell receptor alpha chain (TRAC) and CD52 to prevent graft versus host disease and to render them resistant to anti-CD52 antibody used for lymphodepletion. Preliminary clinical trial data on the use of UCART19 in B-ALL was previously reported at ASH (Benjamin et al, 2018). The phenotypic and functional characteristics of CAR T cell products manufactured from B-ALL, Chronic Lymphocytic Leukaemia (CLL) and DLBCL pts were compared to young adult healthy donor (HD) CAR T cell products. In addition, potential effects related to knocking out TRAC in HD TCR-CAR T cells were examined. Thawed PBMCs from B-ALL, CLL, DLBCL pts and HDs underwent T cell enrichment, activation with anti-CD3/CD28 beads and IL-2, followed by transduction with anti-CD19 4-1BB CD3ζ lentiviral CAR construct and expansion. HD TCR- CAR T cells were manufactured by electroporation of HD CAR T cells with mRNA coding for TRAC TALEN® and residual TCRαβ+cells were removed by magnetic bead selection. CAR expression levels, T cell subsets, and exhaustion markers were examined by flow cytometry. Expression of activation markers CD25 and CD69 was measured in response to co-culture with the CD19+cell line NALM-6. Cytotoxicity against NALM-6 and Raji was assessed and antigen-mediated proliferation measured over 14 days. HD CAR T cells (n=11) expanded significantly more during manufacture than CAR T cells derived from B-ALL (n=9), CLL (n=8) or DLBCL (n=8) pts. As expected, the electroporation step resulted in a transient decrease in viability which recovered over time in culture (n=10). Median CAR expression level was higher on CLL CAR T cell products compared to those from B-ALL pts and HDs, thought to be due to a higher CD4:CD8 ratio in some CLL products. As a consequence of TCR knockout, CD3 expression was lost on HD TCR- CAR T cells (n=10), apart from a small population of γδ CAR T cells. CLL and DLBCL CD8+CAR+cells expressed higher levels of PD1 than HD CD8+CAR+cells. DLBCL CD4+CAR+cells also expressed significantly higher levels of PD1 than HD or HD TCR-CD4+CAR+T cells. CAR+CD8+CD27+PD1- T cells have been previously described as a functionally important population that correlated with clinical outcome in pts who received CLL CAR T cells (Fraietta et al, 2018). We found HD (n=13) and HD TCR- (n=10) CAR T cells had significantly more CD8+CD27+PD1- CAR T cells compared to those derived from CLL (n=8) and DLBCL (n=6) pts, but similar levels to B-ALL pts (n=10). In the absence of CD19 antigen, DLBCL CAR+CD8+ T cells (n=6) had greater expression of CD25 and CD69. However, in response to stimulation with CD19+ NALM-6 cells, HD (n=12), HD TCR- (n=10) and B-ALL (n=10) CAR T cells had higher fold increase in CD69+ cells compared to DLBCL (n=6) CAR T cells. On paired analysis (n=6), no difference was seen in activation in response to CD19 antigen on HD compared to HD TCR- CAR T cells. All CAR T cell products demonstrated comparable cytotoxicity against NALM-6 and Raji cell lines in short term in vitro assays. However, long-term cytotoxicity will be evaluated in a murine model. We performed a detailed comparison of the phenotypic and functional characteristics of CAR T cells derived from pts with B-cell malignancies and HDs. DLBCL CAR T cells showed lower antigen specific activation but higher baseline activation which could lead to more differentiated exhausted T cells. CAR T cells derived from HDs show a higher proportion of the therapeutically relevant CAR+CD8+CD27+PD1- cells compared to patients with mature B cell malignancies (CLL and DLBCL), which is maintained after TRAC knockout. This suggests allogeneic CAR T cells, such as UCART19, may provide a more effective product for pts with T cell dysfunction. Disclosures Graham: Gillead: Other: Funding to attend educational meeting; Servier: Research Funding. Jozwik:Servier: Research Funding. Metelo:Pfizer: Research Funding; Allogene: Research Funding. Almena-Carrasco:Servier: Employment. Peranzoni:Servier: Employment. Ramsay:Celgene Corporation: Research Funding; Roche Glycart AG: Research Funding. Dupouy:Servier: Employment. Farzaneh:Autolus Ltd: Equity Ownership, Research Funding. Patten:Gilead: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Abbvie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen: Consultancy, Honoraria; Roche: Honoraria, Research Funding. Benjamin:Amgen: Honoraria; Allogene: Research Funding; Gilead: Honoraria; Servier: Research Funding; Eusapharm: Consultancy; Pfizer: Research Funding; Takeda: Honoraria; Novartis: Honoraria.
- Published
- 2019
35. Real-World Data of High-Grade Lymphoma Patients Treated with CD19 CAR-T in England
- Author
-
Graham P. Collins, Adrian Bloor, Piers E.M. Patten, Kristian M. Bowles, Maeve A O'Reilly, Rod Johnson, Clare Rowntree, Stephen D. Robinson, R. Sanderson, Muhammad Saif, Kate Robinson, Claire Roddie, Sridhar Chaganti, Ram Malladi, John Radford, Geoff Shenton, Jane Norman, David Irvine, Nuria Martinez-Cibrian, Orla Stewart, Antonio Pagliuca, Andrew McMillan, Tobias Menne, Sanne Lugthart, Arzhang Ardavan, Kim Linton, Andrea Kuhnl, Maria A V Marzolini, and Wendy Osborne
- Subjects
High-grade lymphoma ,Pediatrics ,medicine.medical_specialty ,business.industry ,education ,Immunology ,Disease progression ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Lymphoma ,Transplantation ,medicine ,Marginal zone B-cell lymphoma ,Car t cells ,business ,Diffuse large B-cell lymphoma ,Real world data ,health care economics and organizations - Abstract
Background After European Medicines Agency (EMA) approval of axicabtagene ciloleucel and tisagenlecleucel for the treatment of relapsed/refractory (r/r) high-grade lymphoma in 2018, England was one of the first European countries granting fully funded access to these CD19 CAR-T therapies. Both products are available through the National Health Service England (NHSE) Cancer Drug Fund until their cost-effectiveness has been determined. The NHSE CAR-T program has been set up in a structure aiming to implement robust and transparent criteria for patient selection and to ensure equity of treatment access: CAR-T slots are approved by a weekly National CAR-T Clinical Panel (NCCP), consisting of independent clinical experts, patient representatives, and delegates from each CAR-T centre; treatment is delivered in 7 geographically spread commissioned CAR-T centres (Birmingham, Bristol, King's College Hospital London, University Hospital London, The Christie Manchester, Manchester Royal Infirmary, Newcastle). Here, we report prospective data on the first 122 lymphoma patients approved by the NCCP. Methods Patients with r/r high-grade lymphoma referred to the NCCP between December 2018 and July 2019 and deemed eligible for treatment with CD19 CAR-T were analysed. Eligibility was assessed in the CAR-T centre's tumor board, based on organ function and fitness (performance status 0/1), absence of active CNS disease, and biopsy confirmation of r/r high-grade lymphoma. The final decision on patient eligibility was made by consensus through the NCCP independent clinical panel. CAR-T product selection for each patient was done by the CAR-T centre, mainly on the basis of manufacturing slot availability. Results 122 patients were approved for treatment with CD19 CAR-T therapy by the panel. CAR-T centres selected 76 patients for axicabtagene ciloleucel and 46 for tisagenlecleucel. Patients' median age was 56 years (range 18-75). 62% were male. 87 (71%) patients had de novo diffuse large B-cell lymphoma, 29 (24%) transformed lymphoma (23 from follicular- and 6 from marginal zone lymphoma), and 6 (5%) primary mediastinal B-cell lymphoma. 96 (79%) patients had biopsy confirmation of disease prior to submission. 71 (58%) patients had received 2 prior lines of therapy for high-grade lymphoma, 51 (42%) patients 3 or more treatment lines (maximum 6). 5 patients had previous allogeneic, 19 previous autologous transplant. 88% of patients (107/122) were refractory to the last line of treatment (stable- or progressive disease (PD) or relapse within 6 months). Among 122 patients, 112 completed leukapheresis, 3 are awaiting the procedure, and 7 patients did not proceed (6 due to PD, 1 opted for radical radiotherapy). 57 of 112 patients were infused at the time of abstract submission, 42 are awaiting CAR-T infusion. 10 patients did not proceed to infusion due to disease progression and clinical deterioration (3 with CNS relapse), 2 due to manufacturing failure. One patient achieved a complete response following bridging therapy and is currently monitored. 84% (88/105) patients received bridging therapy between the time of NCCP approval and CAR-T infusion (median 64 days), 62 had chemotherapy, 9 radiotherapy, and 17 steroids only. Details on bridging therapy, treatment-related toxicities and outcomes will be provided at the meeting, by which time approximately 62 patients will have completed their 3 months PET response assessment. Conclusion NHSE has successfully implemented a national structure for providing licenced CAR-T products in England, enabling equity of access and oversight on capacity and patient outcomes, which can serve as a model for newly licenced, cost-intense and complex cell- and gene therapies in the future. The prospective and centralised nature of this dataset offers a true reflection of the real-world patient population undergoing CAR-T therapy in England. Disclosures Kuhnl: Kite Gilead: Honoraria. Roddie:Gilead: Consultancy, Speakers Bureau; Celgene: Consultancy, Speakers Bureau; Novartis: Consultancy. Menne:Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Kite/Gilead: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel grant, Research Funding, Speakers Bureau; Roche: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Bayer: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel grant, Research Funding, Speakers Bureau; Kyowa Kirin: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel grant, Research Funding, Speakers Bureau; Daiichi Sankyo: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel grant, Research Funding, Speakers Bureau; Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Astra Zeneca: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel grant; Jazz: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel grant, Research Funding, Speakers Bureau; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel grant, Research Funding, Speakers Bureau. Sanderson:Kite/Gilead: Honoraria. Osborne:Novartis: Other: Travel; Pfizer: Honoraria, Speakers Bureau; MSD: Consultancy; Takeda: Consultancy, Honoraria, Other: Travel, Speakers Bureau; Roche: Consultancy, Honoraria, Other: Travel, Speakers Bureau; Servier: Consultancy; Gilead: Consultancy. Radford:AstraZeneca: Equity Ownership, Research Funding; Takeda: Consultancy, Honoraria, Research Funding; BMS: Consultancy, Honoraria; Novartis: Consultancy, Honoraria; ADC Therapeutics: Consultancy, Research Funding; GSK: Equity Ownership; Seattle Genetics: Consultancy, Honoraria. Patten:Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Gilead: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Abbvie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen: Consultancy, Honoraria; Roche: Honoraria, Research Funding. O'Reilly:Kite Gilead: Honoraria. Bloor:Abvie, Gilead, Novartis, Autolus, Celgene, etc: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Educational grant. Rowntree:Novartis: Consultancy. Bowles:Abbvie: Research Funding; Janssen: Research Funding. Collins:Gilead: Consultancy, Honoraria. McMillan:BMS: Honoraria; Celgene: Honoraria, Speakers Bureau; F. Hoffmann-La Roche Ltd: Honoraria, Speakers Bureau; Gilead: Honoraria; Novartis: Honoraria; Sandoz: Honoraria; Pfizer: Honoraria, Research Funding; MSD: Honoraria.
- Published
- 2019
36. Abstract CT132: Umbralisib monotherapy demonstrates efficacy and safety in patients with relapsed/refractory marginal zone lymphoma: A multicenter, open-label, registration directed Phase II study
- Author
-
Nathan Fowler, Chan Yoon Cheah, Jeff P. Sharman, Hari P. Miskin, Felipe Samaniego, Nilanjan Ghosh, Peter Sportelli, Pier Luigi Zinzani, Julio C. Chavez, Kathryn S. Kolibaba, Piers E.M. Patten, Paolo Ghia, Ewa Lech-Marańda, Wojciech Jurczak, James A. Reeves, Michael S. Weiss, Owen A. O'Connor, John M. Burke, Corrado Tarella, Bertrand Anz, Piotr Smolewski, and Lori A. Leslie
- Subjects
Cancer Research ,Chemotherapy ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Phases of clinical research ,Neutropenia ,medicine.disease ,Gastroenterology ,Regimen ,Oncology ,Tolerability ,Chemoimmunotherapy ,Internal medicine ,medicine ,Rituximab ,business ,Febrile neutropenia ,medicine.drug - Abstract
Background: Rituximab (RTX) alone or in combination with chemotherapy has substantially improved treatment outcomes for patients (pts) with marginal zone lymphoma (MZL), but relapse is common and not all pts are acceptable candidates for, or respond to, current salvage therapies. Umbralisib is a novel, next-generation PI3K-delta inhibitor with unique inhibition of casein kinase-1ϵ (CK1ϵ) and, compared to earlier generation PI3K-delta inhibitors, exhibits a differentiated tolerability profile with reduced rates of immune-mediated toxicity (Burris et al, 2018). This registration-directed study evaluates the efficacy and safety of umbralisib in pts with relapsed/refractory (R/R) MZL. Methods: Pts had histologically confirmed MZL, ECOG PS ≤2, and had previously received ≥1 prior therapy including at least one CD20 monoclonal antibody (mAb)-containing regimen. All pts received umbralisib 800 mg orally once daily until progression or unacceptable toxicity. The primary study endpoint was overall response rate (ORR) as assessed by an independent review committee (IRC) according to 2007 IWG criteria. ORR by investigator assessment is reported here, and ORR by IRC is forthcoming. Secondary endpoints included duration of response (DOR), progression-free survival (PFS), and safety. Results: Sixty-nine pts were enrolled; we report on the first 38 who are eligible for at least 6 months (mos) of follow-up as of the data cutoff date. Disease status for the 38 pts: extranodal (n=23), nodal (n=8), and splenic (n=7). Median age was 67 years (range, 34-81). Median number of prior systemic therapies was 2 (range, 1-5). Seven pts (18%) had received monotherapy RTX only, and 26 (68%) had received at least one CD20 mAb-containing chemoimmunotherapy. As of the cut-off date, the median follow-up was 9.6 mos. Per investigator assessment, ORR was 55% (4 CRs and 17 PRs), 29% of pts (n=11) had stable disease (SD) of which 6 of these SD pts remain on study with durations ranging from 7-12+ mos. The clinical benefit rate (CR+PR+SD) was 84%, and 91% of pts with at least 1 post-baseline assessment experienced tumor reductions. The median time to initial response was 2.7 mos, while the median DOR was not reached (95% CI: 8.4-not reached). The 12-month PFS was 71%. The most common (≥20%) adverse events (AE) of any grade included: diarrhea (45%), nausea (29%), fatigue (26%), headache (26%), cough (24%), and decreased appetite (21%). The most common Grade 3/4 events were neutropenia (8%), febrile neutropenia (5%), and diarrhea (5%). As of the cutoff date, 16 pts discontinued treatment (PD: 18%; AEs: 8%; pt decision: 8%; physician decision: 8%) and 58% continue treatment. Conclusions: PI3K-delta inhibition with single-agent umbralisib is active and well tolerated in pts with R/R MZL, achieving durable responses with chemotherapy-free therapy. Citation Format: Nathan H. Fowler, Felipe Samaniego, Wojciech Jurczak, Ewa Lech-Maranda, Nilanjan Ghosh, Bertrand Anz, Piers Patten, James A. Reeves, Lori A. Leslie, Piotr Smolewski, Julio C. Chavez, Paolo Ghia, Corrado Tarella, John M. Burke, Jeff Sharman, Kathryn Kolibaba, Owen A. O'Connor, Chan Y. Cheah, Hari P. Miskin, Peter Sportelli, Michael S. Weiss, Pier Luigi Zinzani. Umbralisib monotherapy demonstrates efficacy and safety in patients with relapsed/refractory marginal zone lymphoma: A multicenter, open-label, registration directed Phase II study [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr CT132.
- Published
- 2019
37. IGHV-unmutated and IGHV-mutated chronic lymphocytic leukemia cells produce activation-induced deaminase protein with a full range of biologic functions
- Author
-
Jonathan E. Kolitz, Matthew D. Scharff, Patricia K. A. Mongini, Thomas MacCarthy, Lu Zhang, Kanti R. Rai, Rajendra N. Damle, Sergio Roa, Amanda R. Magli, Steven L. Allen, Piers E.M. Patten, Charles C. Chu, Emilia Albesiano, Xiao-Jie Yan, Dorothy Kim, Jacqueline C. Barrientos, and Nicholas Chiorazzi
- Subjects
Chronic lymphocytic leukemia ,Molecular Sequence Data ,Immunology ,Immunoglobulin Variable Region ,Kaplan-Meier Estimate ,medicine.disease_cause ,Biochemistry ,Somatic evolution in cancer ,Gene Expression Regulation, Enzymologic ,immune system diseases ,Cytidine Deaminase ,Sequence Homology, Nucleic Acid ,hemic and lymphatic diseases ,Tumor Cells, Cultured ,medicine ,Activation-induced (cytidine) deaminase ,Humans ,DNA Breaks, Double-Stranded ,RNA, Messenger ,neoplasms ,Cells, Cultured ,Mutation ,Microscopy, Confocal ,Lymphoid Neoplasia ,Base Sequence ,biology ,Gene Expression Regulation, Leukemic ,Reverse Transcriptase Polymerase Chain Reaction ,Cell Biology ,Hematology ,Cytidine deaminase ,Flow Cytometry ,medicine.disease ,Immunoglobulin Class Switching ,Leukemia, Lymphocytic, Chronic, B-Cell ,Molecular biology ,Leukemia ,Microscopy, Fluorescence ,Immunoglobulin class switching ,Leukocytes, Mononuclear ,biology.protein ,Immunoglobulin Heavy Chains ,IGHV@ ,Cell Division - Abstract
Clonal evolution occurs during the course of chronic lymphocytic leukemia (CLL) and activation-induced deaminase (AID) could influence this process. However, this possibility has been questioned in CLL because the number of circulating AID mRNA+ cells is exceedingly low; synthesis of AID protein by blood CLL cells has not been demonstrated; the full range of AID functions is lacking in unmutated CLL (U-CLL), and no prospective analysis linking AID expression and disease severity has been reported. The results of the present study show that circulating CLL cells and those within secondary lymphoid tissues can make AID mRNA and protein. This production is related to cell division because more AID mRNA was detected in recently divided cells and AID protein was limited to the dividing fraction and was up-regulated on induction of cell division. AID protein was functional because AID+ dividing cells exhibited more double-stranded DNA breaks, IGH class switching, and new IGHV-D-J mutations. Each of these actions was documented in U-CLL and mutated CLL (M-CLL). Furthermore, AID protein was associated with worse patient outcome and adverse cytogenetics. We conclude that the production of fully functional AID protein by U-CLL and M-CLL cells could be involved in clonal evolution of the disease.
- Published
- 2012
38. Phenotype and immune function of lymph node and peripheral blood CLL cells are linked to transendothelial migration
- Author
-
Benedetta Apollonio, Elisabeth Jane Walsby, Piers E.M. Patten, Kirsty Cuthill, Chris Pepper, Linda Barber, Elizabeth H Phillips, Maria Serena Longhi, Christopher Fegan, Eve Coulter, Deborah Yallop, Yun Ma, Andrea G. S. Buggins, Marta Pasikowska, Alan G. Ramsay, and Stephen Devereux
- Subjects
CD4-Positive T-Lymphocytes ,Male ,0301 basic medicine ,Chronic lymphocytic leukemia ,Immunology ,Population ,Biology ,Biochemistry ,03 medical and health sciences ,Immune system ,Antigens, CD ,immune system diseases ,hemic and lymphatic diseases ,medicine ,Humans ,education ,neoplasms ,Lymph node ,CD86 ,education.field_of_study ,Manchester Cancer Research Centre ,ResearchInstitutes_Networks_Beacons/mcrc ,Transendothelial and Transepithelial Migration ,Cell Biology ,Hematology ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Neoplasm Proteins ,030104 developmental biology ,medicine.anatomical_structure ,Lymphatic system ,Cancer research ,Female ,Endothelium, Vascular ,CD80 ,Homing (hematopoietic) - Abstract
Several lines of evidence suggest that homing of tumor cells to lymphoid tissue contributes to disease progression in chronic lymphocytic leukemia (CLL). Here, we demonstrate that lymph node (LN)-derived CLL cells possess a distinct phenotype, and exhibit enhanced capacity for T-cell activation and superior immune synapse formation when compared with paired peripheral blood (PB) samples. LN-derived CLL cells manifest a proliferative, CXCR4(dim)CD5(bright) phenotype compared with those in the PB and higher expression of T-cell activation molecules including CD80, CD86, and HLA-D-related (DR). In addition, LN-CLL cells have higher expression of α4β1 (CD49d) which, as well as being a co-stimulatory molecule, is required for CLL cells to undergo transendothelial migration (TEM) and enter the proliferation centers of the LNs. Using an in vitro system that models circulation and TEM, we showed that the small population of CLL cells that migrate are CXCR4(dim)CD5(bright) with higher CD49d, CD80, CD86, and HLA-DR compared with those that remain circulating; a phenotype strikingly similar to LN-derived CLL cells. Furthermore, sorted CD49d(hi) CLL cells showed an enhanced capacity to activate T cells compared with CD49d(lo) subpopulations from the same patient. Thus, although PB-CLL cells have a reduced capacity to form immune synapses and activate CD4(+) T cells, this was not the case for LN-CLL cells or those with the propensity to undergo TEM. Taken together, our study suggests that CLL cell immunologic function is not only modulated by microenvironmental interactions but is also a feature of a subpopulation of PB-CLL cells that are primed for lymphoid tissue homing and interaction with T cells.
- Published
- 2016
39. ALK-positive large B-cell lymphoma with strong CD30 expression; a diagnostic pitfall and resistance to brentuximab and crizotinib
- Author
-
Deborah Yallop, Piers E.M. Patten, Chirag Shah, Shireen Kassam, Sabine Pomplum, Varun Mehra, Stephen Devereux, Robert Marcus, and Robin M. Ireland
- Subjects
0301 basic medicine ,Histology ,Immunoconjugates ,CD30 ,Pyridines ,Antineoplastic Agents ,Drug resistance ,ALK positive large B-cell lymphoma ,Pathology and Forensic Medicine ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Fatal Outcome ,Crizotinib ,Medicine ,Neoplasm ,Humans ,Diagnostic Errors ,Brentuximab vedotin ,In Situ Hybridization, Fluorescence ,Brentuximab Vedotin ,business.industry ,Large cell ,General Medicine ,medicine.disease ,Lymphoma ,030104 developmental biology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,Lymphoma, Large-Cell, Anaplastic ,Pyrazoles ,Female ,Lymphoma, Large B-Cell, Diffuse ,business ,medicine.drug - Published
- 2016
40. Torque Teno Virus 10 Isolated by Genome Amplification Techniques from a Patient with Concomitant Chronic Lymphocytic Leukemia and Polycythemia Vera
- Author
-
Lu Zhang, Sebastien Didier, Saul Teichberg, Arjun Dhayalan, William J. Kennedy, Jonathan E. Kolitz, Rajendra N. Damle, Charles C. Chu, Amanda R. Magli, Briana M. Agagnina, Prasad Koduru, Gia Fraher, Xiao-Jie Yan, Steven L. Allen, Piers E.M. Patten, Kanti R. Rai, Nicholas Chiorazzi, and Linda P. Johnson
- Subjects
Male ,Torque teno virus ,Chronic lymphocytic leukemia ,Blood Donors ,Genome, Viral ,Biology ,CD38 ,medicine.disease_cause ,Virus ,law.invention ,Polycythemia vera ,law ,hemic and lymphatic diseases ,Genetics ,medicine ,Humans ,Polycythemia Vera ,Molecular Biology ,Genetics (clinical) ,Polymerase chain reaction ,Aged ,Aged, 80 and over ,Mutation ,Reproducibility of Results ,Articles ,Middle Aged ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Virology ,DNA Virus Infections ,Case-Control Studies ,DNA, Viral ,Immunology ,Molecular Medicine ,Female ,IGHV@ ,Nucleic Acid Amplification Techniques - Abstract
An infectious etiology has been proposed for many human cancers, but rarely have specific agents been identified. One difficulty has been the need to propagate cancer cells in vitro to produce the infectious agent in detectable quantity. We hypothesized that genome amplification from small numbers of cells could be adapted to circumvent this difficulty. A patient with concomitant chronic lymphocytic leukemia (CLL) and polycythemia vera (PV) requiring therapeutic phlebotomy donated a large amount of phlebotomized blood to test this possibility. Using genome amplification methods, we identified a new isolate (BIS8-17) of torque teno virus (TTV) 10. The presence of blood isolate sequence 8-17 (BIS8-17) in the original plasma was confirmed by polymerase chain reaction (PCR), validating the approach, since TTV is a known plasma virus. Subsequent PCR testing of plasmas from additional patients showed that BIS8-17 had a similar incidence (~20%) in CLL (n = 48) or PV (n = 10) compared with healthy controls (n = 52). CLL cells do not harbor BIS8-17; PCR did not detect it in CLL peripheral blood genomic deoxyribonucleic acid (DNA) (n = 20). CLL patient clinical outcome or prognostic markers (immunoglobulin heavy chain variable region [IGHV ] mutation, CD38 or zeta-chain associated protein kinase 70 kDa [ZAP-70]) did not correlate with BIS8-17 infection. Although not causative to our knowledge, this is the first reported isolation and detection of TTV in either CLL or PV. TTV could serve as a covirus with another infectious agent or TTV variant with rearranged genetic components that contribute to disease pathogenesis. These results prove that this method identifies infectious agents and provides an experimental methodology to test correlation with disease.
- Published
- 2011
41. Evidence for a macromolecular complex in poor prognosis CLL that contains CD38, CD49d, CD44 and MMP-9
- Author
-
Deborah Yallop, Andrea Pepper née Buggins, K Fishlock, Piers E.M. Patten, Ana Levi, Yolanda Calle, Satyen H. Gohil, and Stephen Devereux
- Subjects
medicine.medical_specialty ,Pathology ,Hematology ,Cell adhesion molecule ,Chronic lymphocytic leukemia ,Cell ,CD44 ,Biology ,CD38 ,Matrix metalloproteinase ,MMP9 ,medicine.disease ,medicine.anatomical_structure ,hemic and lymphatic diseases ,Internal medicine ,medicine ,biology.protein - Abstract
Progressive chronic lymphocytic leukaemia is characterized by the accumulation of neoplastic B-cells in the tissues and correlates with the expression of prognostic biomarkers, such as CD38, CD49d and matrix metalloproteinase-9 (MMP9), which are involved in migration and tissue invasion. In this study we investigated the physical relationship between these molecules and demonstrated that CD38, CD49d, MMP9 and CD44 were physically associated in a supramolecular cell surface complex. Our findings provide a molecular basis for the correlation between expression of these proteins and prognosis and, as the complex is not present in normal B-cells, suggest a novel leukaemia-specific therapeutic target.
- Published
- 2011
42. Ibrutinib Plus Venetoclax in Relapsed/Refractory CLL: Results of the Bloodwise TAP Clarity Study
- Author
-
Stephen Devereux, Christopher P. Fox, Francesco Forconi, Rebecca Bishop, Alison McCaig, Andy C. Rawstron, John G. Gribben, Kristian Brock, Piers E.M. Patten, Talha Munir, Andrew R. Pettitt, Christopher Fegan, Samuel Munoz Vicente, Francesca Yates, Anna Schuh, Peter Hillmen, Donald Macdonald, and Adrian Bloor
- Subjects
medicine.medical_specialty ,Venetoclax ,business.industry ,Immunology ,Refractory CLL ,Cell Biology ,Hematology ,Biochemistry ,Peripheral blood ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,chemistry ,030220 oncology & carcinogenesis ,Family medicine ,Ibrutinib ,Relapsed refractory ,medicine ,Combined therapy ,In patient ,Merck Sharp & Dohme ,business ,030215 immunology - Abstract
BACKGROUND Antigen-mediated proliferation and Bcl-2 mediated survival are key to chronic lymphocytic leukemia (CLL) pathogenesis. Ibrutinib is an oral BTK inhibitor affecting antigen-induced proliferation and cell adhesion/migration whilst venetoclax is a potent, highly selective, orally bioavailable Bcl-2 inhibitor affecting CLL cell survival. The treatment of CLL has been revolutionized by these targeted therapies. Both significantly improve survival in CLL but rarely lead to eradication of detectable minimal residual disease (MRD) when given as single agents. The Bloodwise TAP CLARITY trial combined ibrutinib with venetoclax in order to eradicate detectable CLL with the intention of stopping therapy. METHODS AND PATIENTS CLARITY is a Phase II trial combining ibrutinib with venetoclax in 50 patients with relapsed or refractory CLL. After 8 weeks of ibrutinib monotherapy (420mg/day), venetoclax was added first at a dose of 10mg/day with weekly escalations to 20mg, 50mg, 100mg, 200mg to a final dose of 400mg/day. No tumour lysis syndrome (TLS) was seen for the first 3 patients starting at 10mg/day of venetoclax so all subsequent patients began at 20mg/day. The primary end-point of CLARITY was the eradication of MRD ( RESULTS There were no cases of clinical tumor lysis syndrome (TLS) and only a single case of biochemical TLS (grade 3) which resolved with treatment. Other side-effects were mild and/or manageable, most commonly neutropenia (3/32 grade 2, 29/32 grade 3/4) or gastro-intestinal events (268/278 grade 1/2, 10/278 grade 3/4). Two Suspected Unexpected Serious Adverse Reactions (SUSARs) were reported (abdominal pain and pemphigus), 31 Serious Adverse Events (SAEs), and 918 Adverse Events (AEs) (of which 75 were grade 3 or 4) were reported. Notably there were nine grade 3 or 4 infections and 29 events of grade 3 or 4 neutropenia. Thus far all SAEs have resolved with appropriate management, and all patients remained on trial following resolution. After 6 months of combined ibrutinib plus venetoclax, undetectable MRD was achieved in 19/49 (39%) patients in peripheral blood (PB) and 12/49 (24%) in bone marrow (BM). After 12 months of combined therapy all patients had responded by IWCLL criteria and 23/40 (58%) had achieved a complete remission (CR or CRi). In addition after 12 months of combined therapy 27/31 (87%) have no morphological evidence of CLL in the BM biopsy, 32/34 (94%) had less than 1% CLL cells in the BM aspirate, undetectable MRD (MRD4) was achieved in 23/40 (58%) patients in PB and 17/41 (41%) in BM. There was a continuous improvement in the depth of MRD reduction with 41% of patients achieving less than MRD4 and 29% having undetectable disease by flow cytometry ( CONCLUSIONS The combination of ibrutinib and venetoclax was well tolerated in patients with relapsed or refractory CLL. Every patient responded and there was a high rate of MRD eradication, in some cases leading to the cessation of therapy. Figure Figure. Disclosures Hillmen: Pharmacyclics: Research Funding; Gilead Sciences, Inc.: Honoraria, Research Funding; Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Alexion Pharmaceuticals, Inc: Consultancy, Honoraria; Acerta: Membership on an entity's Board of Directors or advisory committees; Abbvie: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; F. Hoffmann-La Roche Ltd: Research Funding; Celgene: Research Funding; Novartis: Research Funding. Rawstron:Pharmacyclics: Consultancy, Research Funding; Gilead: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Beckman Coulter: Research Funding; BD Biosciences: Research Funding; AbbVie: Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Membership on an entity's Board of Directors or advisory committees, Research Funding; Roche: Membership on an entity's Board of Directors or advisory committees, Research Funding. Brock:GlaxoSmithKline: Equity Ownership; AstraZeneca: Equity Ownership; Lilly: Honoraria; Roche: Other: Reimbursement of expenses; Merck Sharp Dohme: Other: Reimbursement of conference fees. Fegan:Abbvie: Honoraria; Roche: Honoraria; Napp: Honoraria; Gilead Sciences, Inc.: Honoraria; Janssen: Honoraria. McCaig:AbbVie: Membership on an entity's Board of Directors or advisory committees; Gilead: Speakers Bureau. Schuh:Giles, Roche, Janssen, AbbVie: Honoraria. Pettitt:AstraZeneca: Research Funding; Celgene: Research Funding; Gilead: Research Funding; Roche: Research Funding; GSK/Novartis: Research Funding; Napp: Research Funding; Chugai: Research Funding. Gribben:NIH: Research Funding; Novartis: Honoraria; Cancer Research UK: Research Funding; Medical Research Council: Research Funding; TG Therapeutics: Honoraria; Abbvie: Honoraria; Celgene: Consultancy, Honoraria, Research Funding; Janssen: Honoraria, Research Funding; Acerta Pharma: Honoraria, Research Funding; Kite: Honoraria; Unum: Equity Ownership; Wellcome Trust: Research Funding; Pharmacyclics: Honoraria; Roche: Honoraria. Patten:L Hoffman La Roche: Honoraria, Research Funding; Gilead Sciences: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: travel, Research Funding; AbbVie Inc: Honoraria, Other: travel; Janssen: Honoraria, Other: travel. Devereux:Gilead: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Abbvie: Membership on an entity's Board of Directors or advisory committees; Janssen: Other: Personal fees; Novartis: Membership on an entity's Board of Directors or advisory committees. Bloor:AbbVie: Research Funding; Janssen: Research Funding. Fox:Roche: Consultancy, Other: travel support, Research Funding, Speakers Bureau; Sunesis: Consultancy; Gilead: Consultancy, Other: travel support, Research Funding, Speakers Bureau; Celgene: Consultancy, Other: travel support, Speakers Bureau; Abbvie: Consultancy, Other: travel support, Research Funding, Speakers Bureau; Janssen: Consultancy, Other: travel support, Speakers Bureau. Forconi:Janssen-Cilag: Consultancy; Abbvie: Consultancy. Munir:Janssen: Honoraria; Abbvie: Honoraria; Gilead: Honoraria; Novartis: Honoraria; Alexion: Honoraria; MorphoSys: Membership on an entity's Board of Directors or advisory committees.
- Published
- 2018
43. Eliciting Anti-Tumor T Cell Immunity in Chronic Lymphocytic Leukemia (CLL) with PD-L1/PD-1 Blockade Is Enhanced By Avadomide Immunotherapy through the Triggering of Immunogenic Interferon Signaling
- Author
-
Fadi Towfic, Patrick Hagner, Anna Vardi, Anita Gandhi, Nikolaos Ioannou, Kostas Stamatopoulos, Alan G. Ramsay, and Piers E.M. Patten
- Subjects
0301 basic medicine ,business.industry ,medicine.medical_treatment ,Chronic lymphocytic leukemia ,T cell ,Lymphocyte ,Immunology ,Cell Biology ,Hematology ,Immunotherapy ,medicine.disease ,Biochemistry ,Immune checkpoint ,03 medical and health sciences ,030104 developmental biology ,Cytokine ,medicine.anatomical_structure ,Aldesleukin ,medicine ,Cancer research ,Nivolumab ,business - Abstract
Immune checkpoint blockade has demonstrated potential to reactivate anti-tumor immunity and regress tumors. However, response rates in B-cell non-Hodgkin lymphoma patients have been lower compared to Hodgkin lymphoma, with no activity reported in a recent trial of anti-PD-1 immunotherapy in relapsed CLL. This suggests that certain lymphoma subtypes may harbor non-immunogenic tumor microenvironments (TME). Our preliminary studies revealed that avadomide (CC-122), a cereblon modulator, can enhance T cell immune synapse signaling with autologous CLL cells, resulting in a concomitant increase in PD-1/PD-L1 expression at repaired synapses, indicating that avadomide may represent a complementary treatment partner for checkpoint inhibition. Here, we have extended our pre-clinical studies to investigate how these immunotherapy drugs alter the function and gene signatures of previously exhausted T cells from treatment naïve CLL patients (representing disease heterogeneity). Cytotoxicity assays revealed that treating primary T cells and autologous CLL cells with avadomide (1 μM, 48h) activated anti-tumor T cell killing function (P We next performed RNA sequencing on highly purified T cells from treatment naïve CLL patients, representing extremes of prognosis (n=6 good and n=6 poor including disease harboring TP53 abnormalities), following 18 h treatment with avadomide (100 nM) or anti-PD-1 (nivolumab) or PD-L1 (durvalumab) alone (10 μg/ml) or in combinations. Differential expression pathway analysis revealed that the top functional gene categories common for all the avadomide and combination treated samples (independent of anti-PD-1 or anti-PD-L1 monotherapy) were related to the response to type I and II IFN signaling, as well as inflammatory/stimulatory cytokine TNF-α, proliferative IL-6/JAK/STAT3, and IL-2/STAT5 responses. Type I IFN drives expression of chemokines CXCL10 and CXCL9, which are linked to enhanced tumor-infiltrated lymphocyte recruitment, and these chemoattractant genes were significantly upregulated in the avadomide and combination treated patient samples. In addition to their immunostimulatory roles, type II IFN (IFN-γ) and chronic type I IFN signaling have been linked to T cell resistance/exhaustion. We detected upregulated PD-L1 transcript in avadomide and combination treated T cells - supporting blockade of this inhibitory ligand. To investigate the ability of avadomide to modulate T cell migration, we performed comparative quantitative time-lapse microscopy analysis of pretreated patient T cells (n=12). These assays revealed that avadomide, as well as anti-PD-1 or anti-PD-L1 monotherapies, significantly enhanced (P Patient-derived xenograft models demonstrate that the therapeutic treatment of established tumors (3 weeks post-engraftment) with avadomide treatment (0.5 mg/kg) can activate anti-tumor T cells and significantly reduce disease volume (P In conclusion, our findings support the concept that PD-L1/PD-1 blockade in CLL could be enhanced when combined with avadomide through the promotion of immunogenic IFN signaling that enhances T cell infiltration and function. We believe these data support the rationale for combination immunotherapy that could convert a non-immunogenic TME into a 'hot' T cell inflamed TME which would be sensitive to checkpoint blockade. Disclosures Hagner: Celgene Corporation: Employment, Equity Ownership. Towfic:Celgene Corporation: Employment, Equity Ownership. Gandhi:Celgene Corporation: Employment, Equity Ownership. Stamatopoulos:Abbvie: Honoraria, Research Funding; Gilead: Honoraria, Research Funding; Janssen: Honoraria, Research Funding. Patten:L Hoffman La Roche: Honoraria, Research Funding; Gilead Sciences: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: travel, Research Funding; AbbVie Inc: Honoraria, Other: travel; Janssen: Honoraria, Other: travel. Vardi:Gilead: Research Funding; Janssen: Honoraria. Ramsay:MedImmune: Research Funding; Roche Glycart AG: Research Funding; Celgene Corporation: Research Funding.
- Published
- 2018
44. Preliminary Results of UCART19, an Allogeneic Anti-CD19 CAR T-Cell Product, in a First-in-Human Trial (CALM) in Adult Patients with CD19+ Relapsed/Refractory B-Cell Acute Lymphoblastic Leukemia
- Author
-
Victoria Potter, Alan Dunlop, Premal H. Patel, Frédéric Dubois, Charlotte Graham, Agnieszka Jozwik, Florence Binlich, Svetlana Balandraud, Reuben Benjamin, Shireen Kassam, Victoria Metaxa, Stephen Devereux, Sandra Dupouy, Piers E.M. Patten, Anne Philippe, Amina Zinai, Rose Ellard, Cyril Konto, Antonio Pagliuca, Ghulam J. Mufti, Farzin Farzaneh, Deborah Yallop, and Orla Stewart
- Subjects
0301 basic medicine ,medicine.medical_specialty ,business.industry ,Immunology ,Context (language use) ,Cell Biology ,Hematology ,Neutropenia ,medicine.disease ,Biochemistry ,Minimal residual disease ,Fludarabine ,Transplantation ,03 medical and health sciences ,Regimen ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Alemtuzumab ,Rituximab ,business ,medicine.drug - Abstract
Background UCART19 is a genetically modified T-cell product manufactured from non-HLA matched healthy donor cells. Lentiviral-transduced CAR T-cells express (1) an anti-CD19 CAR (anti-CD19 scFv- 41BB- CD3ζ) and (2) an RQR8 "safety switch" that is intended to allow targeted elimination of RQR8+ cells by rituximab. UCART19 has been additionally modified to disrupt the T-cell receptor alpha constant (TRAC) and CD52 genes. The preliminary results of this "off-the-shelf" allogeneic CAR T-cell therapy in a phase I, dose-escalation trial of UCART19 in CD19+ R/R B-ALL adult patients (pts) are described. Methods The primary objective of this study is to determine the maximum tolerated dose of UCART19 by investigating up to four dose levels (DL) in separate sequential cohorts. Adult pts (age ≥16 years) with CD19+ R/R B-ALL who have exhausted available treatment options are eligible. Disease burden must be quantifiable morphologically or with a minimal residual disease (MRD) load ≥1x10-3 at the end of the last anti-leukemic treatment. The lymphodepletion regimen combines cyclophosphamide and fludarabine, with or without alemtuzumab (FC or FCA). A single dose of UCART19 is administered on Day 0, and pts are closely monitored for safety and anti-leukemic activity until the end of study, 3 months after UCART19 administration. Pts are then rolled-over into a 15-years long-term follow-up study. The dose escalation follows a modified Toxicity Probability Interval (mTPI) design based on the occurrence of dose-limiting toxicity (DLT) assessed at the end of the 28-day evaluation period post UCART19 (D28). Results As of 24 June 2017, the 2 first cohorts (3 pts each) who received the first DL (DL1=6x106 total CAR+ cells) have been completed. Median age was 22.5 years (range 18-42). Pts received 1 to 5 previous lines of treatment with 5 out of 6 pts having undergone an allogeneic stem cell transplant (allo-SCT). Four of them had relapsed within 4-6 months post-transplant. Prior to UCART19 infusion, 4 pts had low disease burden ( All pts experienced cytokine release syndrome (CRS): 1 G1, 4 G2 and 1 G4. CRS G1 and G2 were manageable by supportive care ± tocilizumab. CRS G4, assessed as a DLT, occurred in the context of neutropenic sepsis, and was considered to be a contributory factor in the patient's death from multiple organ failure at D15. Time to onset of first CRS symptoms ranged between D5 and D10. CRS correlated with serum cytokine increase (IL-6; IL-10 and INFγ) and UCART19 expansion in the blood. One patient was reported to have probable skin GvHD G1. Only G1 neurotoxic events were observed in 1 patient. Asymptomatic viral reactivations (CMV and/or adenovirus) were seen in 3 pts and resolved with antiviral therapy. Among the 6 pts, 4 achieved a CRi with MRD negativity at D28 (MRD-ve, defined as a tumor burden All 4 pts achieving MRD-ve remission underwent a subsequent allo-SCT, 3 of them within 3 months of UCART19 infusion and 1 following retreatment with FC lymphodepletion and the same dose of UCART19, this patient having relapsed with CD19+ disease 2 months post initial UCART19 infusion. Post allo-SCT, 1 patient relapsed at 100 days with CD19+ disease, 1 died from infection and 2 remain in complete remission. Three pts remain alive at 2.4, 5.3 and 10.2 months respectively post UCART19 treatment. UCART19 (both cells and transgene levels) peaked between D12 and D17 in blood (flow cytometry [figure 1] and qPCR, respectively). UCART19 was detectable in blood from D10 to D28 (up to D42 in 1 patient) and in BM aspirates performed at D14 and D28. In-vivo cell expansion in BM occurred in all but the refractory patient. Conclusion Preliminary results of this first-in-human trial of UCART19 treatment in a high risk R/R B-ALL adult population revealed no unexpected toxicities. Asymptomatic lymphodepletion-related viral reactivations and a probable skin GvHD G1 were encountered. CRi with MRD-ve was achieved in 4 out of 5 pts who reached D28. The 2 first cohorts treated at DL1 have been completed and DL2 will now be investigated on which further results may be presented. The study is active in the UK and will be expanded to other EU countries and the US (NCT 02746952). Disclosures Graham: Servier: Research Funding; Pfizer: Other: Educational meeting attendance; Gilead: Other: Educational meeting attendance; Sanofi: Other: Educational meeting attendance. Yallop: Jazz Pharmaceuticals: Honoraria; Amgen: Honoraria; Pfizer: Other: Advisory board. Jozwik: Servier: Research Funding. Patten: Gilead Inc: Honoraria, Research Funding; Roche: Honoraria; Abbvie: Honoraria. Ellard: Moldmed: Honoraria. Potter: Pfizer: Other: Advisory board; Jazz: Honoraria. Devereux: AbbVie: Consultancy, Honoraria; MSD: Consultancy, Honoraria; Roche: Consultancy, Other: travel expenses; GSK: Consultancy; Gilead: Consultancy, Honoraria, Other: travel expenses, Speakers Bureau; Janssen: Consultancy, Honoraria, Other: travel expenses, Speakers Bureau; Servier: Other: Advisory board. Pagliuca: Jazz: Honoraria; Merck: Honoraria, Research Funding; Bluebird: Honoraria; Pfizer: Honoraria; Basilea: Honoraria; Astellas: Consultancy, Speakers Bureau; Gilead: Honoraria. Zinai: Servier: Employment. Binlich: Servier: Employment. Dupouy: Servier: Employment. Philippe: Servier: Employment. Balandraud: Servier: Employment. Dubois: Servier: Employment. Konto: Bristol-Myers Squibb: Employment, Equity Ownership; Pfizer: Employment, Equity Ownership. Patel: Pfizer: Employment, Equity Ownership. Benjamin: Pfizer: Other: Participated in Adboard meeting, Research Funding; Servier: Research Funding; Celgene: Honoraria.
- Published
- 2017
45. CD38 expression in chronic lymphocytic leukemia is regulated by the tumor microenvironment
- Author
-
Andrew Wotherspoon, Ghulam J. Mufti, Julie Richards, Terry J. Hamblin, Piers E.M. Patten, Andrea G. S. Buggins, Stephen Devereux, and Jon Salisbury
- Subjects
CD4-Positive T-Lymphocytes ,CD31 ,Cell signaling ,Lymphoma ,Endothelium ,Chronic lymphocytic leukemia ,Immunology ,Cell ,Cell Communication ,Biology ,CD38 ,Biochemistry ,immune system diseases ,hemic and lymphatic diseases ,medicine ,Humans ,Tumor microenvironment ,Gene Expression Regulation, Leukemic ,Cell Biology ,Hematology ,medicine.disease ,ADP-ribosyl Cyclase 1 ,Leukemia, Lymphocytic, Chronic, B-Cell ,Leukemia ,medicine.anatomical_structure ,Endothelium, Vascular ,Lymph Nodes - Abstract
Chronic lymphocytic leukemia (CLL) is a lymphoproliferative disease with a highly variable outcome. The prognosis of patients with CLL may be predicted using a number of biomarkers, including the level of CD38 expression at the leukemic cell surface. This study investigates the hypothesis that CD38 expression by CLL cells reflects interactions with nonmalignant cells within pseudofollicles in secondary lymphoid tissue where tumor cell proliferation is thought to occur. CD38 expression is higher in tissues that contain pseudofollicles compared with those that do not. In addition, we show that CD38 expression in CLL is dynamic, changes in response to contact with activated CD4+ T cells, and identifies cells that are primed to proliferate. Finally, we demonstrate close contact between activated CD4+ T cells and proliferating tumor in primary patient tissue. Proliferating tumor cells in lymph nodes express CD38, which is in turn associated with an increased number of CD31+ vascular endothelial cells. Although the factors resulting in colocalization of tumor, T cells, and endothelium remain unclear, the existence of these cellular clusters may provide an explanation for the association between CD38 expression and adverse outcome in CLL and suggests novel therapeutic targets.
- Published
- 2008
46. Tumor-derived IL-6 may contribute to the immunological defect in CLL
- Author
-
Ghulam J. Mufti, Julie Richards, Piers E.M. Patten, N S B Thomas, Stephen Devereux, and Andrea G. S. Buggins
- Subjects
Cancer Research ,Myeloid ,CD40 ,biology ,Interleukin-6 ,T-Lymphocytes ,Chronic lymphocytic leukemia ,Hematology ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Neoplasm Proteins ,Leukemia ,medicine.anatomical_structure ,Immune system ,Oncology ,Immunology ,medicine ,biology.protein ,Humans ,Bone marrow ,Clone (B-cell biology) ,B cell - Abstract
Chronic lymphocytic leukemia (CLL) is an indolent disorder with a highly variable course whose clinical features arise through the accumulation of tumor cells in the bone marrow, blood and secondary lymphoid tissue. While hematopoietic failure may occur in those with advanced disease, for the majority it is immune dysfunction, manifested as susceptibility to infection or autoimmunity, which dominates the clinical picture. Both the disease and its treatment may affect the number of normal cells in the innate and adaptive immune systems, however, it is clear that there are also more subtle qualitative defects which presumably arise either through contact with the expanded neoplastic B cell compartment or because of the secretion of immunomodulatory cytokines. A variety of functional defects in T cells from patients with CLL have been reported including, reduction in activation-induced CD40L expression1 and abnormalities of gene expression affecting the differentiation of CD4 cells and cytoskeletal function and vesicle transport in CD8 cells.2 Although these abnormalities were shown to arise through contact with the malignant clone it is clear that soluble mediators may also play a role, as we have previously shown in myeloid malignancies.3 In the present study, we sought to characterize and identify soluble immunomodulatory factors in CLL since these might lend themselves to therapeutic intervention.
- Published
- 2007
47. APOBEC gene family expression and hallmarks in chronic lymphocytic leukemia
- Author
-
Charles C Chu, Stefano Vergani, Xiao-Jie Yan, Arvind Dhayalan, Piers E.M. Patten, Thomas MacCarthy, Chaohui Yuan, Jacqueline C. Barrientos, Jonathan E. Kolitz, Steven L. Allen, Kanti R. Rai, and Nicholas Chiorazzi
- Subjects
Immunology ,Immunology and Allergy - Abstract
The hallmark activity of APOBEC (apolipoprotein B mRNA editing enzyme, catalytic polypeptide) family of cytidine deaminases, including activation-induced deaminase (AID) and APOBEC3 genes, has been detected in somatic mutation signatures by ultra-deep sequencing of the genomes of many cancers, including chronic lymphocytic leukemia (CLL). The acquisition of these mutations is hypothesized to lead to the progression towards aggressive disease in cancer. To examine this in CLL, we tested if increased APOBEC family member gene expression in CLL cells, as measured by microarray and quantitative real time PCR, correlated with worse patient outcome. Higher levels of AID, APOBEC3B, APOBEC3F and APOBEC3H in CLL cells correlated with worse patient outcome, whereas APOBEC3G did not. Interestingly, higher levels of a truncated splice variant of APOBEC3F tended to correlate with better patient outcome. The expression of truncated APOBEC3F may possibly interfere with APOBEC family member mutation activity. To test mutation activity, CLL cells were activated by transfer into NOD-scid IL2Rγnull mice, a xenograft model of human CLL, and hallmark mutation signatures in the expressed immunoglobulin variable region (IGHV) of CLL cells were analyzed by targeted ultra-deep sequencing. Induced IGHV mutation hallmarks consistent with AID were found. These data support the hypothesis that expression and mutation activity of APOBEC family members, such as AID, in CLL cells could lead to adverse patient consequences.
- Published
- 2017
48. Uncoupling of BCR Internalization and Signaling in Treatment Naive and Btki-Treated Patients with Chronic Lymphocytic Leukemia
- Author
-
Andrea G. S. Buggins, William Townsend, Piers E.M. Patten, Elizabeth H Phillips, Eve Coulter, Stephen Devereux, NI Folarin, Kirsty Cuthill, and Silvia Mele
- Subjects
Endosome ,Chronic lymphocytic leukemia ,media_common.quotation_subject ,Immunology ,breakpoint cluster region ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,hemic and lymphatic diseases ,medicine ,CD5 ,Signal transduction ,Receptor ,Internalization ,Tyrosine kinase ,media_common - Abstract
Ligation of the B-cell receptor (BCR) results in activation of intracellular signaling as well as internalization and processing of ligand/receptor complexes. BCR responsiveness has been shown to vary markedly between patients with chronic lymphocytic leukaemia (CLL) and is linked to prognosis. Despite the central importance of BCR signaling in CLL and the efficacy of drugs that block this pathway, relatively little is known about the capacity of CLL B-cells to internalize ligands that bind to the BCR. In the present study we investigated whether, like normal B-cells, CLL cells can internalize their BCR following stimulation. First, we assessed to what extent this varies between various prognostic subgroups. Second, given that BCR signaling is thought to be more pronounced within lymphoid tissue, we investigated whether internalization varies between different anatomic sites of the same individual. Finally, we examined the effect of agents that inhibit BCR function by comparing BCR expression and internalization in a cohort of patient before and during therapy with Bruton's tyrosine kinase inhibitors (BTKi). BCR internalization was assessed in two ways. First, we used a pH sensitive dye linked to agonistic anti-IgM (pHrodo-αIgM) to detect the uptake and retention of ligand/receptor complexes in acidified endosomes. Second, BCR internalization was assessed directly by measuring the rate of disappearance of surface IgM following ligation by agonistic anti-IgM. An increase in the percentage of cells showing pHrodo fluorescence above control was detected in all CLL cases studied (mean percentage pHrodo-αIgM uptake = 30.2±2.5%, p Similarly, when BCR function within individual CLL patients was examined, we also found that pHrodo-αIgM uptake varied substantially and was maximal in lymph node (LN) derived CLL cells (p=0.03, n=6) and those in the peripheral blood (PB) that express the highest levels of CD5 (p=0.0001, n=26), a marker that is upregulated following BCR activation. In addition, we found that LN CLL cells expressed higher levels of sIgM than those derived from the PB (p=0.03, n=6). This was a surprising finding, as BCR stimulation is thought to occur within LNs, which might be expected to result in down regulated BCR expression. When the level of BCR internalization and accumulation in the endosomes was adjusted for the number of sIgM molecules, we found that BCR internalization and retention was actually more efficient in anergic cases of CLL (defined by lack of ability to mobilize Ca2+ following stimulation with anti-IgM; p=0.0002, n=26). This observation was supported by direct measurement of the rate of BCR endocytosis, which showed a more rapid internalization in anergic B-cells compared to signaling competent and normal B-cells. Similar findings have previously been reported in a murine model of B-cell anergy. Finally, data from BTKi treated CLL patients showed that, 12 months after commencing treatment, CLL B-cells exhibit lower levels of sIgM expression (p=0.02, n=7) and have more efficient BCR internalization than at the outset (p=0.05, n=7). Since low sIgM expression and more efficient BCR internalization is a feature of B-cell anergy, these results suggest that therapy with BTK inhibitors selectively depletes B-cells that are capable of signaling through the BCR and enriches for those displaying features of anergy. Overall our data show that BCR internalization is uncoupled from intracellular signaling in CLL and is most efficient in cells that demonstrate poor downstream BCR signaling or show features of recent BCR stimulation. These observations are similar to those previously reported in anergic B-cells and provide further evidence for ongoing BCR activation and anergy in CLL. Disclosures Patten: Gilead: Research Funding. Devereux:Gilead: Consultancy, Other: Travel, Accommodations, Expenses, Speakers Bureau; Janssen: Consultancy, Other: Travel, Accommodations, Expenses, Speakers Bureau; Roche: Consultancy, Other: Travel, Accommodations, Expenses ; GSK: Consultancy.
- Published
- 2016
49. The Correlation of APOBEC Gene Family Member Expression with Worse CLL Patient Outcome Suggests a Role in CLL Mutational Evolution
- Author
-
Kanti R. Rai, Jonathan E. Kolitz, Charles C. Chu, Arvind Dhayalan, Jacqueline C. Barrientos, Steven L. Allen, Thomas MacCarthy, Nicholas Chiorazzi, Chaohui Yuan, Piers E.M. Patten, and Xiao-Jie Yan
- Subjects
APOBEC ,APOBEC1 ,Immunology ,Cell Biology ,Hematology ,APOBEC-3G Deaminase ,Cytidine deaminase ,Biology ,Biochemistry ,Helsinki declaration ,Germline mutation ,Cancer research ,IGHV@ ,Gene - Abstract
A mutational signature consistent with APOBEC (apolipoprotein B mRNA editing enzyme, catalytic polypeptide) activity has been identified in somatic mutations found in large-scale surveys of ultra-deep sequencing data from many human cancers including chronic lymphocytic leukemia (CLL). APOBEC is a cytidine deaminase family made up of eleven genes, including AID (activation-induced cytidine deaminase) and APOBEC3B, both of which have been implicated in somatic mutation in various cancers, including CLL. These observations have led to the hypothesis that APOBEC cytidine deaminases may be driving somatic mutations leading to the development of more aggressive cancers. Therefore, we examined APOBEC gene family member RNA expression levels in CLL to test for correlations with expression levels and patient outcome. We further examined if CLL cells generated de novo APOBEC family member mutational patterns in the immunoglobulin variable region gene (IGHV) after implantation in a mouse xenograft model of CLL. CLL peripheral blood mononuclear cells (PBMCs) and associated clinical data were collected from patients after informed consent as approved by the Institutional Review Board at the North Shore-Long Island Jewish Health System and in accordance with the Helsinki Declaration. CLL samples were chosen based on availability with no pre-established inclusion/exclusion criteria. CLL RNA expression levels were examined by microarray or quantitative real-time PCR (qPCR). For microarray studies, CLL B cells were purified prior to RNA isolation and acquisition of microarray expression data using Illumina Human WG6 and HT12 bead chips, followed by quantile normalization using GenomeStudio software (Illumina). For qPCR, RNA expression from CLL PBMCs was measured relative to glyceraldehyde 3-phosphate dehydrogenase gene expression by Taqman assay with Roche UPL probes and LightCycler 480. To examine de novo mutations in CLL, the IGHV region was ultra-deep sequenced (Roche 454 FLX system) from human CLL cells recovered from the NOD/Shi-scid,γcnull (NSG) xenograft mouse model of CLL as approved by the Institutional Animal Care and Use Committee at the North Shore-Long Island Jewish Health System. CLL patient (N = 65) RNA expression by microarray showed very low levels of APOBEC1, 2, 3A, 3B, 3D, 4, and AID, modest levels of APOBEC3C and 3H, and high levels of APOBEC3F and 3G. Higher AID expression levels significantly correlated (P To test if CLL cells can acquire de novo mutations indicative of APOBEC gene family member activity, human CLL cells were transferred into NSG mice. After CLL cells proliferated for 4-14 weeks in this xenograft model, the IGHV region was amplified, ultra-deep sequenced, and analyzed for specific mutational characteristics of various APOBEC gene family members. The results of these ongoing analyses will be presented. In conclusion, the expression levels of the APOBEC gene family members AID, APOBEC3B, and potentially APOBEC3F and 3H, correlate with worse patient outcome. These data are consistent with the hypothesis that APOBEC gene family member activity may promote new mutations at sites outside the IG gene loci leading to the evolution of aggressive CLL. Disclosures Barrientos: Pharmacyclics, Celgene, and Genentech: Membership on an entity's Board of Directors or advisory committees; Gilead, Pharmacyclics, and AbbVie: Research Funding.
- Published
- 2015
50. CC-122 Repairs T Cell Activation in Chronic Lymphocytic Leukemia That Results in a Concomitant Increase in PD-1:PD-L1 and CTLA-4 Immune Checkpoint Expression at the Immunological Synapse
- Author
-
Benedetta Apollonio, Patrick Hagner, Mariam Fanous, Piers E.M. Patten, Mohamed-Reda Benmebarek, Anita Gandhi, Michael Pourdehnad, Alan G. Ramsay, and Stephen Devereux
- Subjects
Tumor microenvironment ,business.industry ,T cell ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,Immune checkpoint ,Immunological synapse ,medicine.anatomical_structure ,Immune system ,CTLA-4 ,Cancer research ,Medicine ,business ,CD8 ,B cell - Abstract
Immunomodulatory drugs (IMiDs®) such as lenalidomide and immune checkpoint blockade (ICB) antibodies can enhance autologous anti-tumor T cell immunity and have the potential to elicit durable control of disease in B cell malignancies. These immunotherapies are likely to be most effective when employed in treatment combinations. Thus, the goal of pre-clinical research should be to reveal mechanisms of action (MOA) in the tumor microenvironment (TME) and identify biomarkers to guide development of combination therapy for patients. CC-122 is a novel first-in-class pleiotropic pathway modifier (PPM®) that has potent anti-proliferative, anti-angiogenic and immunomodulatory activities and is currently in Phase I clinical trials for lymphoma and chronic lymphocytic leukemia (CLL). Here, we have utilized the immunological synapse bioassay to examine T cell interactions with CLL tumor cells (modeling anti-tumor T cell responses in the TME) following CC-122 treatment and measure the expression of co-signaling complexes at the synapse. Conjugation assays and confocal imaging were used to visualize intercellular conjugate interactions and F-actin polymerization at the immune synapse between CD4+ and CD8+ T cells and autologous CLL tumor cells pulsed with superantigen (acting as antigen-presenting cells, APCs). Peripheral blood was obtained from treatment naive CLL patients (n=40) representative of disease heterogeneity. Treatment of both purified CLL cells and CD4+ or CD8+ T cells with CC-122 (0.01 - 1 μM for 24h) dramatically enhanced the number of T cells recognizing tumor cells (% conjugation) and increased the formation of F-actin immune synapses (area, μm2) compared to vehicle treated cells (P In conclusion, our results demonstrate for the first time that CC-122 can activate T cell immune synapse signaling against autologous CLL tumor cells and this immunomodulatory capability is more potent than lenalidomide. We further show that CC-122 activation of T cells is associated with enhanced expression of the co-stimulatory receptor ICOS and co-inhibitory checkpoints CTLA-4 and PD-1 at the synapse site. Importantly, our pre-clinical data demonstrates that this regulatory feedback inhibition can be exploited by the addition of anti-PD-L1, anti-PD-1 or anti-CTLA-4 ICB to CC-122 to more optimally stimulate T cell activity against immunosuppressive tumor cells. Disclosures Hagner: Celgene: Employment, Equity Ownership. Pourdehnad:Celgene: Employment. Gandhi:Celgene: Employment, Equity Ownership. Ramsay:MedImmune: Research Funding; Celgene: Research Funding.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.