1
|
Vidal-Manrique M, Nieuwenstein T, Hooijmaijers L, de Jonge P, Djojoatmo M, Jansen J, van der Waart A, Brock R, Dolstra H. IL-15 transpresentation by ovarian cancer cells improves CD34 + progenitor-derived NK cell's anti-tumor functionality. Oncoimmunology 2025; 14:2465010. [PMID: 39960378 PMCID: PMC11834524 DOI: 10.1080/2162402x.2025.2465010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
Ovarian cancer (OC) is the most lethal gynecological malignancy. As high numbers of Natural Killer (NK) cells in ascites associate with improved survival, the adoptive transfer of allogeneic NK cells is an attractive therapeutic strategy. An approach to further improve NK cell expansion and anti-tumor functionality post-infusion includes IL-15 transpresentation (transIL-15), which involves surface expression of the IL-15 cytokine bound to IL-15Rα. However, others have substantiated that systemic administration of ALT/N-803, a soluble molecule mimicking transIL-15, leads to T cell-mediated rejection of the infused allogeneic NK cell product. In addition, whether transIL-15 induce superior expansion and functionality of our hematopoietic progenitor cell-derived NK cells (HPC-NK) remains understudied. Here, we propose to transfect OC cells with IL-15 and IL-15Rα mRNA and evaluate HPC-NK cell stimulation in vitro. Co-transfection of both mRNAs resulted in surface co-expression of both components, thus mimicking the transIL-15. Importantly, co-culture of HPC-NK cells with transIL-15 OC cells resulted in superior proliferation, IFNγ production, cytotoxicity and granzyme B secretion. Furthermore, we observed uptake of IL-15Rα by HPC-NK cells when co-cultured with transIL-15 OC cells, which associates with NK cell long-term proliferation and survival. Superior killing and granzyme B secretion were also observed in transIL-15 OC spheroids. Our results demonstrate that local delivery of IL-15 and IL-15Rα mRNA to OC tumors may be a safer strategy to boost HPC-NK cell therapy of OC through IL-15 transpresentation.
Collapse
Affiliation(s)
- M. Vidal-Manrique
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - T. Nieuwenstein
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - L. Hooijmaijers
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - P.K.J.D. de Jonge
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M. Djojoatmo
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - J. Jansen
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - A.B. van der Waart
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - R. Brock
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Biochemistry, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - H. Dolstra
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
2
|
Wang M, Krueger JB, Gilkey AK, Stelljes EM, Kluesner MG, Pomeroy EJ, Skeate JG, Slipek NJ, Lahr WS, Claudio Vázquez PN, Zhao Y, Bell JB, Clement K, Eaton EJ, Laoharawee K, Chang JW, Webber BR, Moriarity BS. Precision enhancement of CAR-NK cells through non-viral engineering and highly multiplexed base editing. J Immunother Cancer 2025; 13:e009560. [PMID: 40341025 DOI: 10.1136/jitc-2024-009560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Natural killer (NK) cells' unique ability to kill transformed cells expressing stress ligands or lacking major histocompatibility complexes (MHC) has prompted their development for immunotherapy. However, NK cells have demonstrated only moderate responses against cancer in clinical trials. METHODS Advanced genome engineering may thus be used to unlock their full potential. Multiplex genome editing with CRISPR/Cas9 base editors (BEs) has been used to enhance T cell function and has already entered clinical trials but has not been reported in human NK cells. Here, we report the first application of BE in primary NK cells to achieve both loss-of-function and gain-of-function mutations. RESULTS We observed highly efficient single and multiplex base editing, resulting in significantly enhanced NK cell function in vitro and in vivo. Next, we combined multiplex BE with non-viral TcBuster transposon-based integration to generate interleukin-15 armored CD19 chimeric antigen receptor (CAR)-NK cells with significantly improved functionality in a highly suppressive model of Burkitt's lymphoma both in vitro and in vivo. CONCLUSIONS The use of concomitant non-viral transposon engineering with multiplex base editing thus represents a highly versatile and efficient platform to generate CAR-NK products for cell-based immunotherapy and affords the flexibility to tailor multiple gene edits to maximize the effectiveness of the therapy for the cancer type being treated.
Collapse
Affiliation(s)
- Minjing Wang
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Joshua B Krueger
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Alexandria K Gilkey
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Erin M Stelljes
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Mitchell G Kluesner
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Emily J Pomeroy
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Joseph G Skeate
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Nicholas J Slipek
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Walker S Lahr
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Patricia N Claudio Vázquez
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Yueting Zhao
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jason B Bell
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kendell Clement
- Department of Biomedical Informatics, The University of Utah, Salt Lake City, Utah, USA
| | - Ella J Eaton
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kanut Laoharawee
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jae-Woong Chang
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Beau R Webber
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Branden S Moriarity
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
3
|
Lim YJ, Lim MS, Lee JJ, Bae H, Baek YJ, Kim GS, An Y, Kim SK, Yu D. Evaluation of clinical and immunological responses to recombinant canine interleukin-15 therapy in dogs with cancer: A pilot study. Vet Immunol Immunopathol 2025; 283:110923. [PMID: 40203669 DOI: 10.1016/j.vetimm.2025.110923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 03/06/2025] [Accepted: 03/24/2025] [Indexed: 04/11/2025]
Abstract
Interleukin-15 (IL-15) is a pleiotropic cytokine that plays a pivotal role in innate and adaptive immunity. Therefore, it is a promising therapeutic agent for cancer treatment. Despite growing interest in the use of IL-15 as an immunotherapeutic agent, there have been very few reports on its immunological and clinical effects in canine cancers. In this study, we generated recombinant canine IL-15 (rcIL-15) and evaluated its clinical and immunomodulatory effects in combination with metronomic cyclophosphamide in 15 canines with various tumor types. The treatment outcomes were assessed in a prospective clinical trial. Low-dose cyclophosphamide (12.5 mg/m2, PO, SID) was continuously administered for 8 weeks. Starting on day 14, after administering cyclophosphamide, rcIL-15 (20 μg/kg daily) was injected intravenously for 8 days. The disease control rate for combination therapy was 66.6 %, with the most notable partial response accounting for 33.3 % of hematological malignancies. The adverse events were minimal and primarily of grade 1 severity. Moreover, rcIL-15 administration led to significant elevations in anticancer lymphocyte subsets, such as natural killer and cytotoxic T cells, along with increased Ki-67 expression, indicating cellular proliferation. These changes were correlated with improved clinical outcomes. Our findings underscore the therapeutic potential and safety of combining rcIL-15 and metronomic cyclophosphamide for the treatment of various canine cancers.
Collapse
Affiliation(s)
- Y J Lim
- Department of Integrated Life Science and Technology, Kongju National University, Yesan-gun, Chungnam, Republic of Korea; Department of Companion and Laboratory Animal Science, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea.
| | - M S Lim
- Vaxcell-Bio Therapeutics Co., Ltd., Hwasun, Jeollanamdo, Republic of Korea.
| | - J J Lee
- Vaxcell-Bio Therapeutics Co., Ltd., Hwasun, Jeollanamdo, Republic of Korea; Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea.
| | - H Bae
- College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea.
| | - Y J Baek
- Department of Applied, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea.
| | - G S Kim
- Department of Companion and Laboratory Animal Science, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea; Research Institute for Natural Products, Kongju National University, Yesan-gun, Chungnam, Republic of Korea.
| | - Y An
- Department of Applied, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea.
| | - S K Kim
- Department of Integrated Life Science and Technology, Kongju National University, Yesan-gun, Chungnam, Republic of Korea; Department of Companion and Laboratory Animal Science, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea; Vaxcell-Bio Therapeutics Co., Ltd., Hwasun, Jeollanamdo, Republic of Korea; Department of Applied, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea; Research Institute for Natural Products, Kongju National University, Yesan-gun, Chungnam, Republic of Korea.
| | - D Yu
- College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea.
| |
Collapse
|
4
|
Bucher AM, Behrend J, Ehrengut C, Müller L, Emrich T, Schramm D, Akinina A, Kloeckner R, Sieren M, Berkel L, Kuhl C, Sähn MJ, Fink MA, Móré D, Melekh B, Kardas H, Meinel FG, Schön H, Kornemann N, Renz DM, Lubina N, Wollny C, Both M, Watkinson J, Stöcklein S, Mittermeier A, Abaci G, May M, Siegler L, Penzkofer T, Lindholz M, Balzer M, Kim MS, Römer C, Wrede N, Götz S, Breckow J, Borggrefe J, Meyer HJ, Surov A. CT-Defined Pectoralis Muscle Density Predicts 30-Day Mortality in Hospitalized Patients with COVID-19: A Nationwide Multicenter Study. Acad Radiol 2025; 32:2133-2140. [PMID: 39675998 DOI: 10.1016/j.acra.2024.11.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/17/2024]
Abstract
RATIONALE AND OBJECTIVES The prognostic role of computed tomography (CT)-defined skeletal muscle features in COVID-19 is still under investigation. The aim of the present study was to evaluate the prognostic role of CT-defined skeletal muscle area and density in patients with COVID-19 in a multicenter setting. MATERIALS AND METHODS This retrospective study is a part of the German multicenter project RACOON (Radiological Cooperative Network of the COVID-19 pandemic). The acquired sample included 1379 patients, 389 (28.2%) women and 990 (71.8%) men. In each case, chest CT was analyzed and pectoralis muscle area and density were calculated. Data were analyzed by means of descriptive statistics. Group differences were calculated using the Mann-Whitney-U test and Fisher's exact test. Univariable and multivariable logistic regression analyses were performed. RESULTS The 30-day mortality was 17.9%. Using median values as thresholds, low pectoralis muscle density (LPMD) was a strong and independent predictor of 30-day mortality, HR=2.97, 95%-CI: 1.52-5.80, p=0.001. Also in male patients, LPMD predicted independently 30-day mortality, HR=2.96, 95%-CI: 1.42-6.18, p=0.004. In female patients, the analyzed pectoralis muscle parameters did not predict 30-day mortality. For patients under 60 years of age, LPMD was strongly associated with 30-day mortality, HR=2.72, 95%-CI: 1.17;6.30, p=0.019. For patients over 60 years of age, pectoralis muscle parameters could not predict 30-day mortality. CONCLUSION In male patients with COVID-19, low pectoralis muscle density is strongly associated with 30-day mortality and can be used for risk stratification. In female patients with COVID-19, pectoralis muscle parameters cannot predict 30-day mortality.
Collapse
Affiliation(s)
- Andreas Michael Bucher
- Institute of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Frankfurt, Germany (A.M.B., J.B.)
| | - Julius Behrend
- Institute of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Frankfurt, Germany (A.M.B., J.B.)
| | - Constantin Ehrengut
- Department of Radiology, University Hospital of Leipzig, Leipzig, Germany (C.E., H.J.M.)
| | - Lukas Müller
- Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (L.M., T.E.)
| | - Tilman Emrich
- Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (L.M., T.E.)
| | - Dominik Schramm
- Department of Radiology, University Hospital of Halle, Halle, Germany (D.S., A.A.)
| | - Alena Akinina
- Department of Radiology, University Hospital of Halle, Halle, Germany (D.S., A.A.)
| | - Roman Kloeckner
- Department of Radiology, University Hospital Schleswig-Holstein-Campus Luebeck, Lübeck, Germany (R.K., M.S., L.B.)
| | - Malte Sieren
- Department of Radiology, University Hospital Schleswig-Holstein-Campus Luebeck, Lübeck, Germany (R.K., M.S., L.B.)
| | - Lennart Berkel
- Department of Radiology, University Hospital Schleswig-Holstein-Campus Luebeck, Lübeck, Germany (R.K., M.S., L.B.)
| | - Christiane Kuhl
- Department of Diagnostic Interventional Radiology, University Hospital RWTH Aachen, Aachen, Germany (C.K., M.J.S.)
| | - Marwin-Jonathan Sähn
- Department of Diagnostic Interventional Radiology, University Hospital RWTH Aachen, Aachen, Germany (C.K., M.J.S.)
| | - Matthias A Fink
- Clinic for Diagnostic and Interventional Radiology, University Hospital Heidelberg, Heidelberg, Germany (M.A.F., D.M.)
| | - Dorottya Móré
- Clinic for Diagnostic and Interventional Radiology, University Hospital Heidelberg, Heidelberg, Germany (M.A.F., D.M.)
| | - Bohdan Melekh
- Department of Radiology and Nuclear Medicine, University Hospital of Magdeburg, Magdeburg, Germany (B.M., H.K.)
| | - Hakan Kardas
- Department of Radiology and Nuclear Medicine, University Hospital of Magdeburg, Magdeburg, Germany (B.M., H.K.)
| | - Felix G Meinel
- Department of Radiology, University Hospital of Rostock, Rostock, Germany (F.G.M., H.S.)
| | - Hanna Schön
- Department of Radiology, University Hospital of Rostock, Rostock, Germany (F.G.M., H.S.)
| | - Norman Kornemann
- Department of Radiology, Hannover Medical School, Hanover, Germany (N.K., D.M.R.)
| | - Diane Miriam Renz
- Department of Radiology, Hannover Medical School, Hanover, Germany (N.K., D.M.R.)
| | - Nora Lubina
- Clinic for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital of Augsburg, Augsburg, Germany (L.N., W.C.)
| | - Claudia Wollny
- Clinic for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital of Augsburg, Augsburg, Germany (L.N., W.C.)
| | - Marcus Both
- Department of Radiology, University Hospital of Kiel, Kiel, Germany (M.B., J.W.)
| | - Joe Watkinson
- Department of Radiology, University Hospital of Kiel, Kiel, Germany (M.B., J.W.)
| | - Sophia Stöcklein
- Department of Radiology, University Hospital of the Ludwig-Maximilian University Munich, Munich, Germany (S.S., A.M., G.A.)
| | - Andreas Mittermeier
- Department of Radiology, University Hospital of the Ludwig-Maximilian University Munich, Munich, Germany (S.S., A.M., G.A.)
| | - Gizem Abaci
- Department of Radiology, University Hospital of the Ludwig-Maximilian University Munich, Munich, Germany (S.S., A.M., G.A.)
| | - Matthias May
- Department of Radiology, University Hospital of Erlangen, Erlangen, Germany (M.M., L.S.)
| | - Lisa Siegler
- Department of Radiology, University Hospital of Erlangen, Erlangen, Germany (M.M., L.S.)
| | - Tobias Penzkofer
- Department of Radiology, University Hospital of Berlin, Berlin, Germany (T.P., M.L.)
| | - Maximilian Lindholz
- Department of Radiology, University Hospital of Berlin, Berlin, Germany (T.P., M.L.)
| | - Miriam Balzer
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (M.B., M.S.K.)
| | - Moon-Sung Kim
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (M.B., M.S.K.)
| | - Christian Römer
- Clinic for Radiology, University Hospital of Münster, Münster, Germany (C.R., N.W.)
| | - Niklas Wrede
- Clinic for Radiology, University Hospital of Münster, Münster, Germany (C.R., N.W.)
| | - Sophie Götz
- Department of Radiology, University Hospital of Hamburg, Hamburg, Germany (S.G., J.B.)
| | - Julia Breckow
- Department of Radiology, University Hospital of Hamburg, Hamburg, Germany (S.G., J.B.)
| | - Jan Borggrefe
- Institute of Radiology, Neuroradiology and Nuclear Medicine Minden, Ruhr-University-Bochum, Bochum, Germany (J.B., A.S.)
| | - Hans Jonas Meyer
- Department of Radiology, University Hospital of Leipzig, Leipzig, Germany (C.E., H.J.M.)
| | - Alexey Surov
- Institute of Radiology, Neuroradiology and Nuclear Medicine Minden, Ruhr-University-Bochum, Bochum, Germany (J.B., A.S.).
| |
Collapse
|
5
|
Zhang Y, Li T, Liu A, Cheng Y, Meng F, Zhang R, Lao J, Liu Y, Xu N, Ge Y. IL-15/IL-15Rα-secreting bioengineered adipocytes reactivate NK/CD8 + T cells in ovarian and colon cancer ascites. Int J Biol Macromol 2025; 304:140559. [PMID: 39914546 DOI: 10.1016/j.ijbiomac.2025.140559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/26/2025] [Accepted: 01/30/2025] [Indexed: 02/13/2025]
Abstract
Malignant ascites (MA) presents a complex clinical challenge, linked inextricably to poor prognosis, chemoresistance, and metastasis of peritoneal carcinomatosis (PC). However, standard therapeutic approaches for managing or preventing MA secondary to PC remain unavailable. Here we display that a bioengineered adipocyte, encapsulating long-chain fatty acids and concurrently secreting IL-15 and IL-15 receptor α (IL-15Rα), markedly extends the half-life and bioactivity of IL-15. The bioengineered adipocyte consists of an IL-15-P2A-IL-15Rα-T2A-mCherry cDNA sequence stable transfected 3T3-F442A preadipocyte cell line and dcosahexaenoic acid (DHA) are simultaneously encapsulated in the lipid droplets of mature adipocytes, which release it into the MA upon tumor cell-triggered lipolysis. We demonstrate that the bioengineered adipocytes led to specific expansion and activation of NK/CD8+ T cells response to the IL-15/IL-15Rα complex in MA, thereby reversing immuno-suppressive phenotype of ascitic immune cells and enabling them to recognize and attack cancer cells. This synergistic therapeutic strategy exhibits therapeutical manipulation of the ascitic immune cells, restores normal immune functioning, and suppresses cancer cell metastasis and tumor growth in ovarian cancer and colon cancer, all while minimizing systemic adverse effects.
Collapse
Affiliation(s)
- Yuanxin Zhang
- College of Biological and Food Engineering, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, China; Center for Biomedical Research and Innovation, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, China.
| | - Tong Li
- College of Biological and Food Engineering, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, China; Center for Biomedical Research and Innovation, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, China
| | - Aiping Liu
- College of Biological and Food Engineering, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, China; Center for Biomedical Research and Innovation, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, China
| | - Yaqing Cheng
- College of Biological and Food Engineering, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, China; Center for Biomedical Research and Innovation, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, China
| | - Fanwei Meng
- College of Biological and Food Engineering, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, China; Center for Biomedical Research and Innovation, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, China
| | - Renwen Zhang
- College of Biological and Food Engineering, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, China; Center for Biomedical Research and Innovation, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, China
| | - Jun Lao
- College of Biological and Food Engineering, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, China; Center for Biomedical Research and Innovation, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, China
| | - Yihan Liu
- College of Biological and Food Engineering, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, China; Center for Biomedical Research and Innovation, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, China
| | - Ning Xu
- College of Biological and Food Engineering, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, China; Center for Biomedical Research and Innovation, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, China
| | - Yakun Ge
- College of Biological and Food Engineering, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, China; Center for Biomedical Research and Innovation, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, China.
| |
Collapse
|
6
|
Wang Y, Feng Z, Li L, Zhang L. Advances in the role of NK cells in MDS immune dysfunction and antitumor research. Front Immunol 2025; 16:1511616. [PMID: 40103828 PMCID: PMC11913816 DOI: 10.3389/fimmu.2025.1511616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
MDS is a heterogeneous group of myeloid neoplasms originating from hematopoietic stem cells, with a high risk of transformation into acute myeloid leukemia (AML). Natural Killer (NK) cells, crucial for their role in immune surveillance and efficient tumor cell lysis, experience functional impairments due to the complex microenvironment and cytokine dynamics in MDS. This article focuses on the mechanisms of NK cell dysfunction in MDS and the latest strategies to enhance NK cell activity to restore their anti-MDS efficacy, highlighting their key role and potential in MDS therapy.
Collapse
Affiliation(s)
- Yinglong Wang
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Zuxi Feng
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Lijuan Li
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Liansheng Zhang
- Lanzhou University Second Hospital, Lanzhou, China
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
7
|
Naing A, McKean M, Rosen LS, Sommerhalder D, Shaik NM, Wang IM, Le Corre C, Kern KA, Mishra NH, Pal SK. First-in-human phase I study to evaluate safety, tolerability, pharmacokinetics, pharmacodynamics, immunogenicity, and antitumor activity of PF-07209960 in patients with advanced or metastatic solid tumors. ESMO Open 2025; 10:104291. [PMID: 39965362 PMCID: PMC11876874 DOI: 10.1016/j.esmoop.2025.104291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND PF-07209960 is an antibody-cytokine fusion molecule that consists of a single potency-reduced interleukin-15 (IL-15) mutein and a bivalent high-affinity anti-programmed cell death protein 1 (PD-1) full-length IgG. This phase I study (NCT04628780) evaluated the safety, tolerability, pharmacokinetics (PK), pharmacodynamics, and potential clinical benefits of PF-07209960 in patients with selected locally advanced or metastatic solid tumors for whom no standard therapy was available. MATERIALS AND METHODS Escalating doses (1-30 mg) of PF-07209960 were administered subcutaneously once every 2 weeks in 28-day cycles. The primary endpoints included dose-limiting toxicities (DLTs), adverse events (AEs), and laboratory abnormalities. The secondary endpoints included PK, anti-drug antibodies (ADA) and neutralizing antibodies (NAb) against PF-07209960, and tumor response assessed using RECIST version 1.1. RESULTS Thirty-seven patients received treatment with PF-07209960 (1-, 3-, and 10-mg groups, n = 4 each; 15 mg, n = 3; 20 mg, n = 16; 30 mg, n = 6). The median age was 59.0 years (range 31-88 years). Six (22.2%) patients had DLTs. The most frequently reported treatment-related AEs (TRAEs) (≥50%) were general disorders and administration site condition [21 (56.8%)] and skin and subcutaneous tissue disorders [20 (54.1%)]. The most frequently reported grade ≥3 TRAE was anemia [5 (13.5%)]. Two patients with microsatellite-stable colorectal cancer had confirmed partial response, one each from the PF-07209960 20-mg and 30-mg cohorts, with a duration of response of 9.5 and 3 months, respectively. The rate of ADA was 93.9% (31/33), of which 63.6% (21/33) was treatment induced and 30.3% (10/33) was treatment boosted. CONCLUSION PF-07209960 was generally manageable, with potential antitumor activity in some patients.
Collapse
Affiliation(s)
- A Naing
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, USA.
| | - M McKean
- Sarah Cannon Research Institute (SCRI), Nashville, USA
| | - L S Rosen
- UCLA Santa Monica Hematology-Oncology, Santa Monica, USA
| | | | - N M Shaik
- Clinical Pharmacology and Translational Sciences, Pfizer Inc., La Jolla, USA
| | - I-M Wang
- Clinical Pharmacology and Translational Sciences, Pfizer Inc., La Jolla, USA
| | | | | | | | - S K Pal
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, USA
| |
Collapse
|
8
|
Shoushtari AN, Powell DJ. Tumor-Infiltrating Lymphocyte Therapy for Melanoma and Other Solid Tumors: Looking Back, Yet Moving Forward. Transplant Cell Ther 2025; 31:S581-S590. [PMID: 40089327 DOI: 10.1016/j.jtct.2024.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/24/2024] [Indexed: 03/17/2025]
Abstract
Lifileucel, the first solid tumor adoptive tumor infiltrating lymphocyte (TIL) therapy product to receive regulatory approval in advanced melanoma, represents a critical achievement in the pursuit of improving outcomes using cellular therapies in patients with solid tumors. This review traces the development of adoptive TIL therapy from the initial human studies in melanoma, through recent advances in studies of other solid tumors, and previews ongoing and future areas for preclinical and clinical advances to improve upon this novel therapeutic strategy.
Collapse
Affiliation(s)
- Alexander N Shoushtari
- Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York.
| | - Daniel J Powell
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
9
|
Ewendt F, Drewitz F, Althammer M, Eichler C, Brandsch C, Brey S, Winkler TH, Wilkens MR, St-Arnaud R, Kreutz M, Stangl GI. Vitamin D stimulates Il-15 synthesis in rodent muscle. Biochem Biophys Rep 2025; 41:101925. [PMID: 40134939 PMCID: PMC11935148 DOI: 10.1016/j.bbrep.2025.101925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/24/2024] [Accepted: 01/19/2025] [Indexed: 03/27/2025] Open
Abstract
Besides its classical skeletal function, vitamin D plays a critical role in both skeletal muscle and the immune system. Interleukin-15 (IL-15), which is highly expressed, and secreted complexed with its receptor, IL-15Rα, by skeletal muscle, stimulates the development of immune cells and affects myogenesis and muscle mass. However, little is known about possible regulators of this myokine. To test whether vitamin D could be a regulator of muscle IL-15 and IL-15Rα expression, C2C12 myotubes were treated with vitamin D3 metabolites and analysis were performed in gastrocnemius muscles of rats treated with a single intraperitoneal dose of 1,25(OH)2D3. The role of VDR was investigated by siRNA technique in C2C12 myotubes and in gastrocnemius muscles of vitamin D receptor knockout (Vdr-KO) mice. Treatment of C2C12 myotubes with 1,25(OH)2D3 or 25(OH)D3 increased Il-15 gene expression in a dose-dependent manner and 1,25(OH)2D3 also moderately increased the relative Il-15 protein amount. Rats treated with a single dose of 1,25(OH)2D3 demonstrated a higher mRNA abundance of muscle Il-15 than controls. The 1,25(OH)2D3 effect on Il-15 was considerably weaker in C2C12 myotubes treated with Vdr-specific siRNA. Vdr-KO mice showed significantly lower muscle Il-15 mRNA than WT mice. Il-15Ra mRNA and Il-15/Il-15Rα protein abundance were unaffected by 1,25(OH)2D3-treatment or VDR functionality, and Cyp27b1 activity is not required for 25(OH)D3-mediated Il-15 gene expression. The results provide evidence for a regulatory role of hydroxyvitamin D3 metabolites on the Il-15 synthesis in skeletal muscle cells, which is largely mediated by the VDR.
Collapse
Affiliation(s)
- Franz Ewendt
- Martin Luther University Halle-Wittenberg, Institute of Agricultural and Nutritional Sciences, 06120, Halle (Saale), Germany
| | - Fabienne Drewitz
- Martin Luther University Halle-Wittenberg, Institute of Agricultural and Nutritional Sciences, 06120, Halle (Saale), Germany
| | - Michael Althammer
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Cosima Eichler
- Martin Luther University Halle-Wittenberg, Institute of Agricultural and Nutritional Sciences, 06120, Halle (Saale), Germany
| | - Corinna Brandsch
- Martin Luther University Halle-Wittenberg, Institute of Agricultural and Nutritional Sciences, 06120, Halle (Saale), Germany
| | - Stefanie Brey
- Division of Genetics, Department Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - Thomas H. Winkler
- Division of Genetics, Department Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - Mirja R. Wilkens
- Institute of Animal Nutrition, Nutrition Diseases and Dietetics, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - René St-Arnaud
- Shriners Hospitals for Children - Canada and McGill University, Montréal, Quebec, Canada
| | - Marina Kreutz
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Gabriele I. Stangl
- Martin Luther University Halle-Wittenberg, Institute of Agricultural and Nutritional Sciences, 06120, Halle (Saale), Germany
| |
Collapse
|
10
|
Fukushima H, Furusawa A, Okada R, Fujii Y, Choyke PL, Kobayashi H. Antitumor host immunity enhanced by near-infrared photoimmunotherapy. Cancer Sci 2025; 116:572-580. [PMID: 39663860 PMCID: PMC11875768 DOI: 10.1111/cas.16427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024] Open
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) is a novel antitumor therapy that selectively kills cancer cells by NIR light-triggered photochemical reaction of IRDye700DX within Ab-photoabsorber conjugates (APCs). NIR-PIT induces immunogenic cell death, causing immune cell migration between the tumor and tumor-draining lymph nodes, and expanding multiclonal tumor-infiltrating CD8+ T cells. Crucially, the cytotoxic effects of NIR-PIT are limited to cancer cells, sparing immune cells such as antigen-presenting cells and T cells, which are key players in boosting antitumor host immunity. By modifying the Ab used in APC synthesis, NIR-PIT can be repurposed to target and deplete noncancerous immunosuppressive cells including regulatory T cells, myeloid-derived suppressor cells, and cancer-associated fibroblasts in the tumor microenvironment. Immunosuppressive cell targeted NIR-PIT strongly potentiates antitumor host immunity, including the induction of abscopal effects and the development of immune memory. Furthermore, antitumor immune responses and therapeutic efficacy are synergistically enhanced when NIR-PIT is combined with other immune-activating treatments, such as interleukin-15 and immune checkpoint inhibitors. These new findings make NIR-PIT a valuable tool in the evolving landscape of cancer immunotherapy. This review explains the role of NIR-PIT in activating antitumor host immunity.
Collapse
Affiliation(s)
- Hiroshi Fukushima
- Molecular Imaging Branch, Center for Cancer ResearchNational Cancer Institute, NIHBethesdaMarylandUSA
- Department of UrologyInstitute of Science TokyoTokyoJapan
| | - Aki Furusawa
- Molecular Imaging Branch, Center for Cancer ResearchNational Cancer Institute, NIHBethesdaMarylandUSA
| | - Ryuhei Okada
- Molecular Imaging Branch, Center for Cancer ResearchNational Cancer Institute, NIHBethesdaMarylandUSA
- Department of Head and Neck SurgeryInstitute of Science TokyoTokyoJapan
| | - Yasuhisa Fujii
- Department of UrologyInstitute of Science TokyoTokyoJapan
| | - Peter L. Choyke
- Molecular Imaging Branch, Center for Cancer ResearchNational Cancer Institute, NIHBethesdaMarylandUSA
| | - Hisataka Kobayashi
- Molecular Imaging Branch, Center for Cancer ResearchNational Cancer Institute, NIHBethesdaMarylandUSA
| |
Collapse
|
11
|
Li Z, Wrangle J, He K, Sprent J, Rubinstein MP. IL-15: from discovery to FDA approval. J Hematol Oncol 2025; 18:19. [PMID: 39966991 PMCID: PMC11837486 DOI: 10.1186/s13045-025-01664-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Affiliation(s)
- Zihai Li
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, 43210, USA
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA
| | - John Wrangle
- Division of Hematology and Oncology, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Kai He
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, 43210, USA
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Jonathan Sprent
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, 2010, Australia
- St. Vincent's Clinical School, University of New South Wales, Sydney, 1466, Australia
| | - Mark P Rubinstein
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, 43210, USA.
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
12
|
Champiat S, Garralda E, Galvao V, Cassier PA, Gomez-Roca C, Korakis I, Grell P, Naing A, LoRusso P, Mikyskova R, Podzimkova N, Reinis M, Ouali K, Schoenenberger A, Kiemle-Kallee J, Tillmanns S, Sachse R, Moebius U, Spisek R, Bechard D, Jelinkova LP, Adkins I, Marabelle A. Nanrilkefusp alfa (SOT101), an IL-15 receptor βγ superagonist, as a single agent or with anti-PD-1 in patients with advanced cancers. Cell Rep Med 2025; 6:101967. [PMID: 39933529 PMCID: PMC11866505 DOI: 10.1016/j.xcrm.2025.101967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 10/15/2024] [Accepted: 01/16/2025] [Indexed: 02/13/2025]
Abstract
Nanrilkefusp alfa (nanril; SOT101) is an interleukin (IL)-15 receptor βγ superagonist that stimulates natural killer (NK) and CD8+ T cells, thereby promoting an innate and adaptive anti-tumor inflammatory microenvironment in mouse tumor models either in monotherapy or combined with an anti-programmed cell death protein 1 (PD-1) antibody. In cynomolgus monkeys, a clinical schedule was identified, which translated into the design of a phase 1/1b clinical trial, AURELIO-03 (NCT04234113). In 51 patients with advanced/metastatic solid tumors, nanril increased the proportions of CD8+ T cells and NK cells in peripheral blood and tumors. It had a favorable safety profile when administered subcutaneously on days 1, 2, 8, and 9 of each 21-day cycle as monotherapy (0.25-15 μg/kg) or combined (1.5-12 μg/kg) with the anti-PD-1 pembrolizumab (200 mg). The most frequent treatment-emergent adverse events were pyrexia, injection site reactions, and chills. Furthermore, early clinical efficacy was observed, including in immune checkpoint blockade-resistant/refractory patients.
Collapse
Affiliation(s)
- Stephane Champiat
- Gustave Roussy, Departement d'Innovation Therapeutique et d'Essais Precoces (DITEP), Universite Paris Saclay, 94805 Villejuif, France.
| | - Elena Garralda
- Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | | | | | - Carlos Gomez-Roca
- Institut Universitaire du Cancer de Toulouse, 31100 Toulouse, France
| | - Iphigenie Korakis
- Institut Universitaire du Cancer de Toulouse, 31100 Toulouse, France
| | - Peter Grell
- Masaryk Memorial Cancer Institute, 602 00 Brno, Czech Republic
| | - Aung Naing
- Department of Lnvestigational Cancer Therapeutics, MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Romana Mikyskova
- Laboratory of Immunological and Tumor Models, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | | | - Milan Reinis
- Laboratory of Immunological and Tumor Models, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Kaissa Ouali
- Gustave Roussy, Departement d'Innovation Therapeutique et d'Essais Precoces (DITEP), Universite Paris Saclay, 94805 Villejuif, France
| | | | | | | | | | | | - Radek Spisek
- SOTIO Biotech a.s., 170 00 Prague, Czech Republic
| | | | - Lenka Palova Jelinkova
- SOTIO Biotech a.s., 170 00 Prague, Czech Republic; Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, 150 06 Prague, Czech Republic
| | - Irena Adkins
- SOTIO Biotech a.s., 170 00 Prague, Czech Republic; Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, 150 06 Prague, Czech Republic
| | - Aurelien Marabelle
- Gustave Roussy, Departement d'Innovation Therapeutique et d'Essais Precoces (DITEP), Universite Paris Saclay, 94805 Villejuif, France
| |
Collapse
|
13
|
Guo P, Zhong L, Wang T, Luo W, Zhou A, Cao D. NK cell-based immunotherapy for hepatocellular carcinoma: Challenges and opportunities. Scand J Immunol 2025; 101:e13433. [PMID: 39934640 DOI: 10.1111/sji.13433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 12/22/2024] [Accepted: 01/01/2025] [Indexed: 02/13/2025]
Abstract
Hepatocellular carcinoma (HCC) remains one of the most challenging malignancies globally, characterized by significant heterogeneity, late-stage diagnosis, and resistance to treatment. In recent years, the advent of immune-checkpoint blockades (ICBs) and targeted immune cell therapies has marked a substantial advancement in HCC treatment. However, the clinical efficacy of these existing therapies is still limited, highlighting the urgent need for new breakthroughs. Natural killer (NK) cells, a subset of the innate lymphoid cell family, have shown unique advantages in the anti-tumour response. With increasing evidence suggesting the crucial role of dysfunctional NK cells in the pathogenesis and progression of HCC, considerable efforts have been directed toward exploring NK cells as a potential therapeutic target for HCC. In this review, we will provide an overview of the role of NK cells in normal liver immunity and in HCC, followed by a detailed discussion of various NK cell-based immunotherapies and their potential applications in HCC treatment.
Collapse
Affiliation(s)
- Pei Guo
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Liyuan Zhong
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Tao Wang
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Weijia Luo
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Aiqiang Zhou
- Guangzhou Hospital of Integrated Chinese and Western Medicine, Guangzhou, Guangdong, P.R China
| | - Deliang Cao
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
14
|
Zha B, Ma Y, Zha B, Guo X. Predictive value of temporal muscle thickness for prognosis in newly diagnosed IDH wild-type glioblastoma patients: evaluated for a Chinese population. Eur Radiol 2025:10.1007/s00330-025-11394-7. [PMID: 39884988 DOI: 10.1007/s00330-025-11394-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 12/26/2024] [Accepted: 01/04/2025] [Indexed: 02/01/2025]
Abstract
OBJECTIVES This study aims to investigate the prognostic value of Temporal Muscle Thickness (TMT) in Chinese patients with newly diagnosed isocitrate dehydrogenase (IDH) wild-type glioblastoma. METHODS Data were retrospectively collected from patients with isocitrate dehydrogenase wild-type genotype glioblastoma, who underwent surgical treatment and concurrent chemoradiotherapy at our center between May 2019 and May 2023. Multi-model and multivariate Cox regression were used to examine factors associated with overall and progression-free survival. Subgroup analysis and sensitivity tests were performed to verify the robustness of the results. Restricted cubic spline regression was used to explore the possible nonlinear relationship between TMT and OS/PFS. RESULTS A total of 344 patients were enrolled in this study. The main analysis showed that TMT was positively correlated with overall survival and progression-free survival. The results of multivariate Cox regression after segmentation according to the optimal cutoff value showed that age ≥ 60 years (HR = 1.47, 95% CI: 1.14-1.90, p = 0.003), diabetes (HR = 1.95, 95% CI: 1.26-3.04, p = 0.003) were the risk factors for death. However, TMT ≥ 8.425 mm (HR = 0.45, 95% CI: 0.36-0.57, p < 0.001), Karnofsky ≥ 70 (HR = 0.76, 95% CI: 0.59-0.97, p < 0.031) were associated with a lower risk of death. Age ≥ 60 years (HR = 1.33, 95% CI: 1.03-1.71, p = 0.028) was a risk factor for recurrence, Karnofsky ≥ 70 (HR = 0.76, 95% CI: 0.59-0.97, p = 0.027), TMT ≥ 8.4 mm (HR = 0.64, 95% CI: 0.50-0.80, p < 0.001), and combination of targeted therapy (HR = 0.65, 95% CI: 0.47-0.90, p = 0.01) reduced recurrence risk. CONCLUSION TMT is an independent predictor of OS and PFS in IDH wild-type GBM patients and can be used to predict prognosis in clinical practice. KEY POINTS Question Performance status of glioblastoma patients is crucial for treatment decision-making and prognosis assessment; however, there is currently no objective evaluation metric for the Chinese population. Findings Temporal muscle thickness at initial diagnosis is positively correlated with overall survival and progression-free survival in Chinese patients with newly diagnosed IDH wild-type glioblastoma. Clinical relevance Temporal muscle thickness at initial diagnosis is an independent, objective prognostic factor for newly diagnosed IDH wild-type glioblastoma patients in China. It contributes to the formulation of individualized treatment plans and serves as a stratification factor in clinical trials.
Collapse
Affiliation(s)
- Boya Zha
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yajing Ma
- Department of Neurointervention, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Boqun Zha
- Xinxiang Medical University, Xinxiang, China
| | - Xinbin Guo
- Department of Neurointervention, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
15
|
Jiang H, Limsuwannarot S, Kulhanek KR, Pal A, Rysavy LW, Su L, Labiad O, Testa S, Ogana H, Waghray D, Tao P, Jude KM, Seet CS, Crooks GM, Moding EJ, Garcia KC, Kalbasi A. IL-9 as a naturally orthogonal cytokine with optimal JAK/STAT signaling for engineered T cell therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633105. [PMID: 39868284 PMCID: PMC11760723 DOI: 10.1101/2025.01.15.633105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Arming T cells with a synthetically orthogonal IL-9 receptor (o9R) permits facile engraftment and potent anti-tumor functions. We considered whether the paucity of natural IL-9R expression could be exploited for T cell immunotherapy given that, in mice, high doses of IL-9 were well-tolerated without discernible immune modulation. Compared to o9R, T cells engineered with IL-9R exhibit superior tissue infiltration, stemness, and anti-tumor activity. These qualities are consistent with a stronger JAK/STAT signal, which in addition to STAT1/3/5, unexpectedly includes STAT4 (canonically associated with IL-12 but not common γ-chain cytokines). IL-9R T cells are exquisitely sensitive to perturbations of proximal signaling, including structure-guided attenuation, amplification, and rebalancing of JAK/STAT signals. Biased IL-9R mutants uncover STAT1 as a rheostat between proliferative stem-like and terminally differentiated effector states. In summary, we identify native IL-9/IL-9R as a natural cytokine-receptor pair with near-orthogonal qualities and an optimal JAK/STAT signaling profile for engineered T cell therapy.
Collapse
Affiliation(s)
- Hua Jiang
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA, 94305, USA
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine. Stanford, CA, 94305, USA
- These authors contributed equally to this work
| | - Sam Limsuwannarot
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA, 94305, USA
- These authors contributed equally to this work
| | - Kayla R. Kulhanek
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA, 94305, USA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, 94143, USA
- Stanford Center for Cancer Cell Therapy
- These authors contributed equally to this work
| | - Aastha Pal
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA, 94305, USA
- These authors contributed equally to this work
| | - Lea W. Rysavy
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA, 94305, USA
- These authors contributed equally to this work
| | - Leon Su
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine. Stanford, CA, 94305, USA
| | - Ossama Labiad
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA, 94305, USA
| | - Stefano Testa
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA, 94305, USA
| | - Heather Ogana
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA, 94305, USA
| | - Deepa Waghray
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine. Stanford, CA, 94305, USA
| | - Pingdong Tao
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine. Stanford, CA, 94305, USA
| | - Kevin M. Jude
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine. Stanford, CA, 94305, USA
| | - Christopher S. Seet
- Broad Stem Cell Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Gay M. Crooks
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Division of Pediatric Hematology-Oncology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Everett J. Moding
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA, 94305, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, 94143, USA
| | - K. Christopher Garcia
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine. Stanford, CA, 94305, USA
- Parker Institute for Cancer Immunotherapy, 1 Letterman Drive, Suite D3500, San Francisco, CA 94129, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Anusha Kalbasi
- Department of Radiation Oncology, Stanford University School of Medicine. Stanford, CA, 94305, USA
- Stanford Center for Cancer Cell Therapy
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, 94143, USA
- Lead contact
| |
Collapse
|
16
|
Kureshi CT, Dougan SK. Cytokines in cancer. Cancer Cell 2025; 43:15-35. [PMID: 39672170 PMCID: PMC11841838 DOI: 10.1016/j.ccell.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 12/15/2024]
Abstract
Cytokines are proteins used by immune cells to communicate with each other and with cells in their environment. The pleiotropic effects of cytokine networks are determined by which cells express cytokines and which cells express cytokine receptors, with downstream outcomes that can differ based on cell type and environmental cues. Certain cytokines, such as interferon (IFN)-γ, have been clearly linked to anti-tumor immunity, while others, such as the innate inflammatory cytokines, promote oncogenesis. Here we provide an overview of the functional roles of cytokines in the tumor microenvironment. Although we have a sophisticated understanding of cytokine networks, therapeutically targeting cytokine pathways in cancer has been challenging. We discuss current progress in cytokine blockade, cytokine-based therapies, and engineered cytokine therapeutics as emerging cancer treatments of interest.
Collapse
Affiliation(s)
- Courtney T Kureshi
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Program in Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Stephanie K Dougan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Program in Immunology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
17
|
Steffin D, Ghatwai N, Montalbano A, Rathi P, Courtney AN, Arnett AB, Fleurence J, Sweidan R, Wang T, Zhang H, Masand P, Maris JM, Martinez D, Pogoriler J, Varadarajan N, Thakkar SG, Lyon D, Lapteva N, Zhuyong M, Patel K, Lopez-Terrada D, Ramos CA, Lulla P, Armaghany T, Grilley BJ, Gottschalk S, Dotti G, Metelitsa LS, Heslop HE, Brenner MK, Sumazin P, Heczey A. Interleukin-15-armoured GPC3 CAR T cells for patients with solid cancers. Nature 2025; 637:940-946. [PMID: 39604730 DOI: 10.1038/s41586-024-08261-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024]
Abstract
Interleukin-15 (IL-15) promotes the survival of T lymphocytes and enhances the antitumour properties of chimeric antigen receptor (CAR) T cells in preclinical models of solid neoplasms in which CAR T cells have limited efficacy1-4. Glypican-3 (GPC3) is expressed in a group of solid cancers5-10, and here we report the evaluation in humans of the effects of IL-15 co-expression on GPC3-expressing CAR T cells (hereafter GPC3 CAR T cells). Cohort 1 patients ( NCT02905188 and NCT02932956 ) received GPC3 CAR T cells, which were safe but produced no objective antitumour responses and reached peak expansion at 2 weeks. Cohort 2 patients ( NCT05103631 and NCT04377932 ) received GPC3 CAR T cells that co-expressed IL-15 (15.CAR), which mediated significantly increased cell expansion and induced a disease control rate of 66% and antitumour response rate of 33%. Infusion of 15.CAR T cells was associated with increased incidence of cytokine release syndrome, which was controlled with IL-1/IL-6 blockade or rapidly ameliorated by activation of the inducible caspase 9 safety switch. Compared with non-responders, tumour-infiltrating 15.CAR T cells from responders showed repression of SWI/SNF epigenetic regulators and upregulation of FOS and JUN family members, as well as of genes related to type I interferon signalling. Collectively, these results demonstrate that IL-15 increases the expansion, intratumoural survival and antitumour activity of GPC3 CAR T cells in patients.
Collapse
Affiliation(s)
- David Steffin
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Nisha Ghatwai
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Antonino Montalbano
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Purva Rathi
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Amy N Courtney
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Azlann B Arnett
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
- Department of Immunology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Julien Fleurence
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Ramy Sweidan
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Tao Wang
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Huimin Zhang
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Prakash Masand
- Department of Radiology, Baylor College of Medicine, Houston, TX, USA
| | - John M Maris
- Department of Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Martinez
- Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer Pogoriler
- Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Navin Varadarajan
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Sachin G Thakkar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Deborah Lyon
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Natalia Lapteva
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
- Pathology and Immunology Graduate Program, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Baylor College of Medicine, Houston, TX, USA
| | - Mei Zhuyong
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Kalyani Patel
- Department of Pathology, Baylor College of Medicine, Houston, TX, USA
| | | | - Carlos A Ramos
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Premal Lulla
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Tannaz Armaghany
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Bambi J Grilley
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Leonid S Metelitsa
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Helen E Heslop
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Malcolm K Brenner
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | - Pavel Sumazin
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Andras Heczey
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
- Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX, USA.
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX, USA.
- Texas Children's Hospital Liver Tumor Program, Houston, TX, USA.
| |
Collapse
|
18
|
Han J, Wang H. Cytokine-overexpressing dendritic cells for cancer immunotherapy. Exp Mol Med 2024; 56:2559-2568. [PMID: 39617785 DOI: 10.1038/s12276-024-01353-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/11/2024] [Indexed: 12/28/2024] Open
Abstract
Dendritic cells (DCs), the main type of antigen-presenting cells in the body, act as key mediators of adaptive immunity by sampling antigens from diseased cells for the subsequent priming of antigen-specific T and B cells. While DCs can secrete a diverse array of cytokines that profoundly shape the immune milieu, exogenous cytokines are often needed to maintain the survival, proliferation, and differentiation of DCs, T cells, and B cells. However, conventional cytokine therapies for cancer treatment are limited by their low therapeutic benefit and severe side effects. The overexpression of cytokines in DCs, followed by paracrine release or membrane display, has emerged as a viable approach for controlling the exposure of cytokines to interacting DCs and T/B cells. This approach can potentially reduce the necessary dose of cytokines and associated side effects to achieve comparable or enhanced antitumor efficacy. Various strategies have been developed to enable the overexpression or chemical conjugation of cytokines on DCs for the subsequent modulation of DC-T/B-cell interactions. This review provides a brief overview of strategies that enable the overexpression of cytokines in or on DCs via genetic engineering or chemical modification methods and discusses the promise of cytokine-overexpressing DCs for the development of new-generation cancer immunotherapy.
Collapse
Affiliation(s)
- Joonsu Han
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Cancer Center at Illinois (CCIL), Urbana, IL, USA.
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Carle College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
19
|
Merino A, Brunstein CC, Shanley R, Rashid F, Wangen R, Bachanova V, Juckett M, Maakaron J, Felices M, Weisdorf D, Miller JS. N-803, an IL-15 Superagonist Complex as Maintenance Therapy After Allogeneic Donor Stem Cell Transplant for Acute Myeloid Leukemia or Myelodysplastic Syndrome; A Phase 2 Trial. Transplant Cell Ther 2024; 30:1206.e1-1206.e12. [PMID: 39362494 PMCID: PMC11887273 DOI: 10.1016/j.jtct.2024.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 10/05/2024]
Abstract
Maintenance therapy may improve natural killer (NK) cell surveillance after allogeneic donor hematopoietic cell transplant (HCT) for myeloid malignancies and represents a potential approach to improve cure rates. Interleukin-15 (IL-15) enhances lymphocyte proliferation and antitumor activity. In a prior Phase 1 study of an IL-15 superagonist (N-803) in patients with AML who relapsed after HCT, we observed in vivo expansion of NK cells and antitumor responses. The primary objective of this Phase 2 trial was to determine if post-transplant N-803 could reduce relapse. We administered N-803 (n = 20) (dosed 6 mcg/kg subcutaneously [SQ] at day 60 after HCT to patients with myelodysplastic syndrome [MDS] or acute myeloid leukemia [AML] who were in complete remission [CR]). N-803 treatment was planned weekly, biweekly or every 4 weeks in 2 sequential cohorts. The most common adverse events after administration were self-limited injection sites skin rashes (n = 20). One week after an N-803 dose, we observed enhanced NK cell proliferation and improved antitumor cytotoxicity without inducing immune exhaustion. Five patients who developed acute graft versus host disease (aGVHD) after N-803 responded promptly to steroids and 4 patients developed chronic GVHD. Patients receiving >4 doses of N-803 had a 3-fold decrease in relapse at two years (P = .06). These findings support the safety, immune activation, and potential efficacy of N-803 to prevent relapse of AML/MDS after HSCT.
Collapse
Affiliation(s)
- Aimee Merino
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, Minnesota.
| | | | - Ryan Shanley
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Faridullah Rashid
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Rose Wangen
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Veronika Bachanova
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Mark Juckett
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Joseph Maakaron
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Martin Felices
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Daniel Weisdorf
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Jeffrey S Miller
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
20
|
Qiao W, Dong P, Chen H, Zhang J. Advances in Induced Pluripotent Stem Cell-Derived Natural Killer Cell Therapy. Cells 2024; 13:1976. [PMID: 39682724 PMCID: PMC11640743 DOI: 10.3390/cells13231976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Natural killer (NK) cells are cytotoxic lymphocytes of the innate immune system capable of killing virus-infected cells and/or cancer cells. The commonly used NK cells for therapeutic applications include primary NK cells and immortalized NK cell lines. However, primary NK cell therapy faces limitations due to its restricted proliferation capacity and challenges in stable storage. Meanwhile, the immortalized NK-92 cell line requires irradiation prior to infusion, which reduces its cytotoxic activity, providing a ready-made alternative and overcoming these bottlenecks. Recent improvements in differentiation protocols for iPSC-derived NK cells have facilitated the clinical production of iPSC-NK cells. Moreover, iPSC-NK cells can be genetically modified to enhance tumor targeting and improve the expansion and persistence of iPSC-NK cells, thereby achieving more robust antitumor efficacy. This paper focuses on the differentiation-protocols efforts of iPSC-derived NK cells and the latest progress in iPSC-NK cell therapy. Additionally, we discuss the current challenges faced by iPSC-NK cells and provide an outlook on future applications and developments.
Collapse
Affiliation(s)
- Wenhua Qiao
- CAMS Key Laboratory for T Cell and Immunotherapy, State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China;
| | - Peng Dong
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou 213000, China;
| | - Hui Chen
- CAMS Key Laboratory for T Cell and Immunotherapy, State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China;
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou 213000, China;
| | - Jianmin Zhang
- CAMS Key Laboratory for T Cell and Immunotherapy, State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China;
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou 213000, China;
| |
Collapse
|
21
|
Chen P, Li S, Nagaoka K, Kakimi K, Kataoka K, Cabral H. Nanoenabled IL-15 Superagonist via Conditionally Stabilized Protein-Protein Interactions Eradicates Solid Tumors by Precise Immunomodulation. J Am Chem Soc 2024; 146:32431-32444. [PMID: 39356776 PMCID: PMC11613988 DOI: 10.1021/jacs.4c08327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
Protein complexes are crucial structures that control many biological processes. Harnessing these structures could be valuable for therapeutic therapy. However, their instability and short lifespans need to be addressed for effective use. Here, we propose an innovative approach based on a functional polymeric cloak that coordinately anchors different domains of protein complexes and assembles them into a stabilized nanoformulation. As the polymer-protein association in the cloak is pH sensitive, the nanoformulation also allows targeting the release of the protein complexes to the acidic microenvironment of tumors for aiding their therapeutic performance. Building on this strategy, we developed an IL-15 nanosuperagonist (Nano-SA) by encapsulating the interleukin-15 (IL-15)/IL-15 Receptor α (IL-15Rα) complex (IL-15cx) for fostering synergistic transpresentation in tumors. Upon intravenous administration, Nano-SA stably circulated in the bloodstream, safeguarding the integrity of IL-15cx until reaching the tumor site, where it selectively released the active complex. Thus, Nano-SA significantly amplified the antitumor immune signals while diminishing systemic off-target effects. In murine colon cancer models, Nano-SA achieved potent immunotherapeutic effects, eradicating tumors without adverse side effects. These findings highlight the transformative potential of nanotechnology for advancing protein complex-based therapies.
Collapse
Affiliation(s)
- Pengwen Chen
- Department
of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Shangwei Li
- Department
of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Koji Nagaoka
- Department
of Immunotherapeutics, The University of
Tokyo Hospital, 7-3-1
Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kazuhiro Kakimi
- Department
of Immunotherapeutics, The University of
Tokyo Hospital, 7-3-1
Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kazunori Kataoka
- Innovation
Center of NanoMedicine (iCONM), Kawasaki
Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Horacio Cabral
- Department
of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
22
|
Sun Z, Xu A, Wu Z, Lan X, Gao G, Guo B, Yu Z, Shao L, Wu H, Lv M, Wang Y, Zhao Y, Wang B. Effect of hypoxia-induced mIL15 expression on expansion and memory progenitor stem-like TILs in vitro. Front Immunol 2024; 15:1450245. [PMID: 39650651 PMCID: PMC11621077 DOI: 10.3389/fimmu.2024.1450245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 11/01/2024] [Indexed: 12/11/2024] Open
Abstract
Introduction The adoptive cell transfer of tumor-infiltrating lymphocytes (TILs) has proven clinically beneficial in patients with non-small cell lung cancer refractory to checkpoint blockade immunotherapy, which has prompted interest in TIL-adoptive cell transfer. The transgenic expression of IL15 can promote the expansion, survival, and function of T cells ex vivo and in vivo and enhance their anti-tumor activity. The effect of expressing mIL15 regulated by hypoxia in the tumor microenvironment on the expansion, survival, and stem-like properties of TILs has not been explored. Methods Using TILs expanded from the tumor tissues of lung cancer patients, TILs with or without mIL15 expression (TIL-mIL15 or UN-TIL) were generated by lentiviral transduction. To reflect the advantages of mTIL15, the cells were divided into groups with IL2 (TIL-mIL15+IL2) or without IL2 (TIL-mIL15-IL2). Results Compared to UN-TIL cells, mIL15 expression had a similar capacity for promoting TIL proliferation and maintaining cell viability. Our experimental findings indicate that, compared to UN-TIL and TIL-mIL15+IL2 cells, the expression of mIL15 in TIL-mIL15-IL2 cells promoted the formation of stem-like TILs (CD8+CD39-CD69-) and led to significant decreases in the proportion and absolute number of terminally differentiated TILs (CD8+CD39+CD69+). RNA-Seq data revealed that in TIL-mIL15-IL2 cells, the expression of genes related to T cell differentiation and effector function, including PRDM1, ID2, EOMES, IFNG, GZMB, and TNF, were significantly decreased, whereas the expression of the memory stem-like T cell marker TCF7 was significantly increased. Furthermore, compared to UN-TIL and TIL-mIL15+IL2 cells, TIL-mIL15-IL2 cells showed significantly lower expression levels of inhibitory receptors LAG3, TIGIT, and TIM3, which was consistent with the RNA-Seq results. Discussion This study demonstrates the superior persistence of TIL-mIL15-IL2 cells, which may serve as a novel treatment strategy for lung cancer patients.
Collapse
Affiliation(s)
- Zhen Sun
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
- Department of Research and Development, Qingdao Sino-Cell Biomed Co., Ltd., Qingdao, Shandong, China
| | - Aotian Xu
- Department of Research and Development, Qingdao Sino-Cell Biomed Co., Ltd., Qingdao, Shandong, China
| | - Zhaojun Wu
- Department of Research and Development, Qingdao Sino-Cell Biomed Co., Ltd., Qingdao, Shandong, China
| | - Xiaohao Lan
- Department of Research and Development, Qingdao Sino-Cell Biomed Co., Ltd., Qingdao, Shandong, China
| | - Ganchen Gao
- Department of Research and Development, Qingdao Sino-Cell Biomed Co., Ltd., Qingdao, Shandong, China
| | - Bin Guo
- Department of Research and Development, Qingdao Sino-Cell Biomed Co., Ltd., Qingdao, Shandong, China
| | - Zhongjie Yu
- Department of Research and Development, Qingdao Sino-Cell Biomed Co., Ltd., Qingdao, Shandong, China
| | - Lin Shao
- Department of Research and Development, Qingdao Sino-Cell Biomed Co., Ltd., Qingdao, Shandong, China
| | - Hao Wu
- Department of Research and Development, Qingdao Sino-Cell Biomed Co., Ltd., Qingdao, Shandong, China
| | - Min Lv
- Department of Research and Development, Qingdao Sino-Cell Biomed Co., Ltd., Qingdao, Shandong, China
| | - Yongjie Wang
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yi Zhao
- Department of Research and Development, Qingdao Sino-Cell Biomed Co., Ltd., Qingdao, Shandong, China
| | - Bin Wang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
- Department of Research and Development, Qingdao Sino-Cell Biomed Co., Ltd., Qingdao, Shandong, China
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
23
|
Portugal I, Clavijo-Salomon MA. Complete response to fifth-line anti-PD-1 rechallenge in fumarate hydratase-mutated papillary renal cell carcinoma. NPJ Precis Oncol 2024; 8:251. [PMID: 39496729 PMCID: PMC11535469 DOI: 10.1038/s41698-024-00750-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 10/28/2024] [Indexed: 11/06/2024] Open
Abstract
Fumarate hydratase (FH) mutated papillary renal cell carcinoma is an aggressive variant of kidney cancer that poorly responds to conventional targeted therapies and immunotherapy. Here, we present the 10-year follow-up of a heavily pre-treated patient with several lines of therapy, achieving a remarkable complete response to anti-PD-1 rechallenge. In addition, we highlight a common immune-related adverse event of anti-PD-1, eosinophilia, as a possible biomarker of response and using TCGA data analysis, provide proof-of-concept for tumor expression of the eosinophil-related gene SIGLEC8, as a promising powerful predictor of prognosis for papillary renal cell carcinoma patients.
Collapse
Affiliation(s)
- Isabella Portugal
- Ciências Médicas, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
- Centro de Investigação Translacional em Oncologia (CTO), Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
- Department of Medicine, Cambridge Health Alliance, Cambridge, MA, USA
| | - Maria A Clavijo-Salomon
- Centro de Investigação Translacional em Oncologia (CTO), Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil.
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil.
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
24
|
Hangasky JA, Fernández RDV, Stellas D, Hails G, Karaliota S, Ashley GW, Felber BK, Pavlakis GN, Santi DV. Leveraging long-acting IL-15 agonists for intratumoral delivery and enhanced antimetastatic activity. Front Immunol 2024; 15:1458145. [PMID: 39559362 PMCID: PMC11570272 DOI: 10.3389/fimmu.2024.1458145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/14/2024] [Indexed: 11/20/2024] Open
Abstract
Introduction IL-15 agonists hold promise as immunotherapeutics due to their ability to induce the proliferation and expansion of cytotoxic immune cells including natural killer (NK) and CD8+ T cells. However, they generally have short half-lives that necessitate frequent administration to achieve efficacy. To address this limitation, we have developed a half-life extension technology using hydrogel microspheres (MS). Here, the therapeutic is tethered to MSs by a releasable linker with pre-programed cleavage rates. We previously showed the MS conjugate of single-chain IL-15, MS~IL-15, effectively increased the half-life of IL-15 to approximately 1 week and enhanced the pharmacodynamics. We sought to determine whether the same would be true with a MS conjugate of the IL-15 agonist, receptor-linker IL-15 (RLI). Methods We prepared a long acting MS conjugate of RLI, MS~RLI. The pharmacokinetics and pharmacodynamics of MS~RLI were measured in C57BL/6J mice and compared to MS~IL-15. The antitumor efficacy of MS~RLI was measured when delivered subcutaneously or intratumorally in the CT26 tumor model and intratumorally in the orthotopic EO771 tumor model. Results MS~RLI exhibited a half-life of 30 h, longer than most IL-15 agonists but shorter than MS~IL-15. The shorter than expected half-life of MS~RLI was shown to be due to target-mediated-disposition caused by an IL-15 induced cytokine sink. MS~RLI resulted in very potent stimulation of NK and CD44hiCD8+ T cells, but also caused significant injection-site toxicity that may preclude subcutaneous administration. We thus pivoted our efforts toward studying the MS~RLI for long-acting intra-tumoral therapy, where some degree of necrosis might be beneficial. When delivered intra- tumorally, both MS~IL-15 and MS~RLI had modest anti-tumor efficacy, but high anti- metastatic activity. Conclusion Intra-tumoral MS~RLI and MS~RLI combined with systemic treatment with other agents could provide beneficial antitumor and anti-metastatic effects without the toxic effects of systemic IL-15 agonists. Our findings demonstrate that intra-tumorally administered long-acting IL-15 agonists counter two criticisms of loco-regional therapy: the necessity for frequent injections and the challenge of managing metastases.
Collapse
Affiliation(s)
| | | | - Dimitris Stellas
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | | | - Sevasti Karaliota
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
- Basic Science Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | | | - Barbara K. Felber
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - George N. Pavlakis
- Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | | |
Collapse
|
25
|
Herrero Alvarez N, Molvi Z, Lupo K, Urraca J, Balderes P, Nyakatura EK, Khan AG, Viray T, Lewis JS, O'Reilly RJ. 89Zr-immunoPET-guided selection of a CD33xIL15 fusion protein optimized for antitumor immune cell activation and in vivo tumour retention in acute myeloid leukaemia. Eur J Nucl Med Mol Imaging 2024; 51:4001-4014. [PMID: 38987489 PMCID: PMC11921880 DOI: 10.1007/s00259-024-06814-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 06/16/2024] [Indexed: 07/12/2024]
Abstract
PURPOSE Immune cells are capable of eliminating leukemic cells, as evidenced by outcomes in hematopoietic cell transplantation (HCT). However, patients who fail induction therapy will not benefit from HCT due to their minimal residual disease (MRD) status. Thus, we aimed to develop an immunomodulatory agent to reduce MRD by activating immune effector cells in the presence of leukaemia cells via a novel fusion protein that chimerises two clinically tolerated biologics: a CD33 antibody and the IL15Ra/IL15 complex (CD33xIL15). METHODS We generated a set of CD33xIL15 fusion protein constructs with varying configurations and identified those with the best in vitro AML-binding, T cell activation, and NK cell potentiation. Using 89Zr-immunoPET imaging we then evaluated the biodistribution and in vivo tumour retention of the most favourable CD33xIL15 constructs in an AML xenograft model. Ex vivo biodistribution studies were used to confirm the pharmacokinetics of the constructs. RESULTS Two of the generated fusion proteins, CD33xIL15 (N72D) and CD33xIL15wt, demonstrated optimal in vitro behaviour and were further evaluated in vivo. These studies revealed that the CD33xIL15wt candidate was capable of being retained in the tumour for as long as its parental CD33 antibody, Lintuzumab (13.9 ± 3.1%ID/g vs 18.6 ± 1.1%ID/g at 120 h). CONCLUSION This work demonstrates that CD33xIL15 fusion proteins are capable of targeting leukemic cells and stimulating local T cells in vitro and of concentrating in the tumour in AML xenografts. It also highlights the importance of 89Zr-immunoPET to guide the development and selection of tumour-targeted antibody-cytokine fusion proteins.
Collapse
Affiliation(s)
- Natalia Herrero Alvarez
- Department of Radiology and Program in Pharmacology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Zaki Molvi
- Immunology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Kyle Lupo
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Jessica Urraca
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Paul Balderes
- Tri-Institutional Therapeutics Discovery Institute, 1230 York Avenue, New York, NY, 10065, USA
| | - Elisabeth K Nyakatura
- Tri-Institutional Therapeutics Discovery Institute, 1230 York Avenue, New York, NY, 10065, USA
| | - Abdul G Khan
- Tri-Institutional Therapeutics Discovery Institute, 1230 York Avenue, New York, NY, 10065, USA
| | - Tara Viray
- Department of Radiology and Program in Pharmacology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Jason S Lewis
- Department of Radiology and Program in Pharmacology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
- Departments of Pharmacology and Radiology, Weill Cornell Medicine, New York, NY, 10065, USA.
| | - Richard J O'Reilly
- Immunology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
26
|
Zhu A, Bai Y, Nan Y, Ju D. Natural killer cell engagers: From bi-specific to tri-specific and tetra-specific engagers for enhanced cancer immunotherapy. Clin Transl Med 2024; 14:e70046. [PMID: 39472273 PMCID: PMC11521791 DOI: 10.1002/ctm2.70046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/25/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024] Open
Abstract
Natural killer cell engagers (NKCEs) are a specialised subset of antibodies capable of simultaneously targeting endogenous NK cells and tumour cells, generating precise and effective cytolytic responses against cancer. This review systematically explores NK engagers as a rising star in NK-mediated immunotherapy, specifically focusing on multi-specific engagers. It examines the diverse configuration of NKCEs and how certain biologics could be employed to boost NK activity, including activating receptor engagement and cytokine incorporation. Some challenges and future perspectives of current NKCEs therapy are also discussed, including optimising pharmacokinetics, addressing the immunosuppressive tumour microenvironment and exploring potential combinatorial approaches. By offering an in-depth analysis of the current landscape and future trajectories of multi-specific NKCEs in cancer treatment, this review serves as a valuable resource for understanding this promising field of immunotherapy. HIGHLIGHTS Innovative NKCEs: NK cell engagers (NKCEs) represent a promising new class of immunotherapeutics targeting tumours by activating NK cells. Multi-specific formats: The transition from bi-specific to multi-specific NKCEs enhances their versatility and therapeutic efficacy. MECHANISMS OF ACTION NKCEs have the potential to improve NK cell activation by engaging activating receptors and incorporating cytokines. CLINICAL POTENTIAL Current clinical trials demonstrate the safety and efficacy of various NKCEs across different cancer types. Future research directions: Optimising NKCE designs and exploring combination therapies are essential for overcoming challenges in cancer treatment.
Collapse
Affiliation(s)
- An Zhu
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunotherapeuticsFudan University School of PharmacyShanghaiChina
- Shanghai Institute of Infectious Disease and BiosecurityFudan UniversityShanghaiChina
| | - Yu Bai
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunotherapeuticsFudan University School of PharmacyShanghaiChina
| | - Yanyang Nan
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunotherapeuticsFudan University School of PharmacyShanghaiChina
| | - Dianwen Ju
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunotherapeuticsFudan University School of PharmacyShanghaiChina
- Shanghai Institute of Infectious Disease and BiosecurityFudan UniversityShanghaiChina
| |
Collapse
|
27
|
Zeng Q, Zeng S, Dai X, Ding Y, Huang C, Ruan R, Xiong J, Tang X, Deng J. MDM2 inhibitors in cancer immunotherapy: Current status and perspective. Genes Dis 2024; 11:101279. [PMID: 39263534 PMCID: PMC11388719 DOI: 10.1016/j.gendis.2024.101279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/13/2024] [Accepted: 02/21/2024] [Indexed: 09/13/2024] Open
Abstract
Murine double minute 2 (MDM2) plays an essential role in the cell cycle, apoptosis, DNA repair, and oncogene activation through p53-dependent and p53-independent signaling pathways. Several preclinical studies have shown that MDM2 is involved in tumor immune evasion. Therefore, MDM2-based regulation of tumor cell-intrinsic immunoregulation and the immune microenvironment has attracted increasing research attention. In recent years, immune checkpoint inhibitors targeting PD-1/PD-L1 have been widely used in the clinic. However, the effectiveness of a single agent is only approximately 20%-40%, which may be related to primary and secondary drug resistance caused by the dysregulation of oncoproteins. Here, we reviewed the role of MDM2 in regulating the immune microenvironment, tumor immune evasion, and hyperprogression during immunotherapy. In addition, we summarized preclinical and clinical findings on the use of MDM2 inhibitors in combination with immunotherapy in tumors with MDM2 overexpression or amplification. The results reveal that the inhibition of MDM2 could be a promising strategy for enhancing immunotherapy.
Collapse
Affiliation(s)
- Qinru Zeng
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
| | - Shaocheng Zeng
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
| | - Xiaofeng Dai
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
| | - Yun Ding
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
| | - Chunye Huang
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
| | - Ruiwen Ruan
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
| | - Xiaomei Tang
- Department of Oncology, Jiangxi Chest Hospital, Nanchang, Jiangxi 330006, China
| | - Jun Deng
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi 330006, China
- Postdoctoral Innovation Practice Base, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| |
Collapse
|
28
|
Chaoul N, Lauricella E, Giglio A, D'Angelo G, Ganini C, Cives M, Porta C. The future of cellular therapy for the treatment of renal cell carcinoma. Expert Opin Biol Ther 2024; 24:1245-1259. [PMID: 39485013 DOI: 10.1080/14712598.2024.2418321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/26/2024] [Accepted: 10/15/2024] [Indexed: 11/03/2024]
Abstract
INTRODUCTION Systemic treatment options for renal cell carcinoma (RCC) have expanded considerably in recent years, and both tyrosine kinase inhibitors and immune checkpoint inhibitors, alone or in combination, have entered the clinical arena. Adoptive cell immunotherapies have recently revolutionized the treatment of cancer and hold the promise to further advance the treatment of RCC. AREAS COVERED In this review, we summarize the latest preclinical and clinical development in the field of adoptive cell immunotherapy for the treatment of RCC, focusing on lymphokine-activated killer (LAK) cells, cytokine-induced killer (CIK) cells, tumor-infiltrating T cells (TILs), TCR-engineered T cells, chimeric antigen receptor (CAR) T cells, and dendritic cell vaccination strategies. Perspectives on emerging cellular products including CAR NK cells, CAR macrophages, as well as γδ T cells are also included. EXPERT OPINION So far, areas of greater therapeutic success of adoptive cell therapies include the adjuvant administration of CIK cells and the transfer of anti-CD70 CAR T cells in patients with metastatic RCC. Bench to bedside and back research will be needed to overcome current limitations of adoptive cell therapies in RCC, primarily aiming at improving the safety of immune cell products, optimizing their antitumor activity and generating off-the-shelf products ready for clinical use.
Collapse
Affiliation(s)
- Nada Chaoul
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Eleonora Lauricella
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Andrea Giglio
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Gabriella D'Angelo
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Carlo Ganini
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy
| | - Mauro Cives
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy
| | - Camillo Porta
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy
| |
Collapse
|
29
|
Qi Z, Gu J, Qu L, Shi X, He Z, Sun J, Tan L, Sun M. Advancements of engineered live oncolytic biotherapeutics (microbe/virus/cells): Preclinical research and clinical progress. J Control Release 2024; 375:209-235. [PMID: 39244159 DOI: 10.1016/j.jconrel.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/26/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
The proven efficacy of immunotherapy in fighting tumors has been firmly established, heralding a new era in harnessing both the innate and adaptive immune systems for cancer treatment. Despite its promise, challenges such as inefficient delivery, insufficient tumor penetration, and considerable potential toxicity of immunomodulatory agents have impeded the advancement of immunotherapies. Recent endeavors in the realm of tumor prophylaxis and management have highlighted the use of living biological entities, including bacteria, oncolytic viruses, and immune cells, as a vanguard for an innovative class of live biotherapeutic products (LBPs). These LBPs are gaining recognition for their inherent ability to target tumors. However, these LBPs must contend with significant barriers, including robust immune clearance mechanisms, cytotoxicity and other in vivo adverse effects. Priority must be placed on enhancing their safety and therapeutic indices. This review consolidates the latest preclinical research and clinical progress pertaining to the exploitation of engineered biologics, spanning bacteria, oncolytic viruses, immune cells, and summarizes their integration with combination therapies aimed at circumventing current clinical impasses. Additionally, the prospective utilities and inherent challenges of the biotherapeutics are deliberated, with the objective of accelerating their clinical application in the foreseeable future.
Collapse
Affiliation(s)
- Zhengzhuo Qi
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Junmou Gu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lihang Qu
- The 4th People's Hospital of Shenyang, China Medical University, Shenyang, Liaoning, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Lingchen Tan
- School of Life Sciences and Biopharmaceutical, Shenyang Pharmaceutical University, Shenyang, Liaoning, China.
| | - Mengchi Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China.
| |
Collapse
|
30
|
Huang F, Ma M, Lang L, Yang S, Zhao H, Zhang J, Liu H. Impact of sarcopenia on the prognosis of patients with advanced non-small cell lung cancer treated with antiangiogenic therapy: A propensity score matching analysis. Thorac Cancer 2024; 15:2248-2259. [PMID: 39308019 PMCID: PMC11543275 DOI: 10.1111/1759-7714.15443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/31/2024] [Accepted: 08/22/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND Limited information is available regarding the impact of sarcopenia on the prognosis of antiangiogenic therapy in individuals with advanced non-small cell lung cancer (NSCLC). This study primarily sought to examine the prognostic significance of sarcopenia in individuals with advanced NSCLC undergoing antiangiogenic therapy. METHODS We retrospectively enrolled all patients who met the inclusion and exclusion criteria from 2019 to 2021 at Nantong University Hospital. Patients were grouped according to the presence or absence of sarcopenia. After propensity score matching (PSM), progression-free survival (PFS), overall survival (OS), and adverse event rates were compared between the two groups. Factors associated with prognosis were screened using univariate and multivariate analyses. RESULTS A total of 267 patients were included, with a total of 201 matched at baseline after PSM (77 in the sarcopenia group and 124 in the non-sarcopenia group). The sarcopenia group had lower PFS (p = 0.043) and OS (p = 0.011) than the non-sarcopenia group and a higher incidence of adverse events (p = 0.044). Multivariate analysis suggested that sarcopenia is an independent prognostic risk factor for OS in advanced NSCLC patients receiving antiangiogenic therapies (p = 0.009). Results of subgroup analyses showed some differences in the impact of sarcopenia on survival prognosis in populations with different characteristics. CONCLUSION Patients with advanced NSCLC with comorbid sarcopenia exhibit a worse prognosis when treated with antiangiogenic therapy, and preventing and ameliorating sarcopenia may lead to better survival outcomes in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Fuchun Huang
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongChina
| | - Mingxuan Ma
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongChina
| | - Liye Lang
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongChina
| | - Shuang Yang
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongChina
| | - Hui Zhao
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongChina
| | - Jialin Zhang
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongChina
| | - Hua Liu
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongChina
| |
Collapse
|
31
|
Lockhart J, Lulla P. "Lymphorepletion" by IL-15. Blood 2024; 144:1650-1652. [PMID: 39418032 DOI: 10.1182/blood.2024025793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
|
32
|
Srinagesh H, Jackson C, Shiraz P, Jeyakumar N, Hamilton M, Egeler E, Mavroukakis S, Kuo A, Cancilla J, Sahaf B, Agarwal N, Kanegai A, Kramer AM, Arai S, Bharadwaj S, Dahiya S, Hosoya H, Johnston L, Kennedy V, Liedtke M, Lowsky R, Mikkilineni L, Negrin R, Rezvani A, Sidana S, Shizuru J, Smith M, Weng WK, Feldman S, Frank MJ, Lee Z, Tagliaferri M, Marcondes AM, Miklos D, Mackall C, Muffly L. A phase 1 clinical trial of NKTR-255 with CD19-22 CAR T-cell therapy for refractory B-cell acute lymphoblastic leukemia. Blood 2024; 144:1689-1698. [PMID: 38968138 PMCID: PMC11522888 DOI: 10.1182/blood.2024024952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 07/07/2024] Open
Abstract
ABSTRACT Although chimeric antigen receptor (CAR) T-cell (CAR-T) therapy has revolutionized the treatment of B-cell malignancies, many patients relapse and therefore strategies to improve antitumor immunity are needed. We previously designed a novel autologous bispecific CAR targeting CD19 and CD22 (CAR19-22), which was well tolerated and associated with high response rates but relapse was common. Interleukin-15 (IL15) induces proliferation of diverse immune cells and can augment lymphocyte trafficking. Here, we report the results of a phase 1 clinical trial of the first combination of a novel recombinant polymer-conjugated IL15 receptor agonist (NKTR-255), with CAR19-22, in adults with relapsed/refractory B-cell acute lymphoblastic leukemia. Eleven patients were enrolled, 9 of whom successfully received CAR19-22 followed by NKTR-255. There were no dose-limiting toxicities, with transient fever and myelosuppression as the most common possibly related toxicities. We observed favorable efficacy with 8 of 9 patients (89%) achieving measurable residual disease-negative remission. At 12 months, progression-free survival for NKTR-255 was double that of historical controls (67% vs 38%). We performed correlative analyses to investigate the effects of IL15 receptor agonism. Cytokine profiling showed significant increases in IL15 and the chemokines CXCL9 and CXCL10. The increase in chemokines was associated with decreases in absolute lymphocyte counts and CD8+ CAR T cells in the blood and 10-fold increases in cerebrospinal fluid CAR-T cells, suggesting lymphocyte trafficking to tissue. Combining NKTR-255 with CAR19-22 was safe, feasible, and associated with high rates of durable responses. This trial was registered at www.clinicaltrials.gov as #NCT03233854.
Collapse
Affiliation(s)
| | - Clayton Jackson
- Division of Hematology/Oncology, UT Southwestern, Dallas, TX
| | - Parveen Shiraz
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | | | - Mark Hamilton
- Division of Hematology, Stanford University, Stanford, CA
| | - Emily Egeler
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | | | - Adam Kuo
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | | | - Bita Sahaf
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | - Neha Agarwal
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Alyssa Kanegai
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Anne Marijn Kramer
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | - Sally Arai
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Sushma Bharadwaj
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Saurabh Dahiya
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Hitomi Hosoya
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Laura Johnston
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Vanessa Kennedy
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | | | - Robert Lowsky
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Lekha Mikkilineni
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Robert Negrin
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Andrew Rezvani
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Surbhi Sidana
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Judith Shizuru
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Melody Smith
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Wen-Kai Weng
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
| | - Steven Feldman
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | - Matthew J. Frank
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | | | | | | | - David Miklos
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | - Crystal Mackall
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| | - Lori Muffly
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA
- Center for Cancer Cell Therapy, Stanford University, Stanford, CA
| |
Collapse
|
33
|
Song YC, Chen SY, Zhao XR, Jing H, Fang H, Tang Y, Hu SY, Song YW, Jin J, Liu YP, Qi SN, Sun GY, Zhong QZ, Du XH, Liu J, Li YX, Wang SL. Prognostic value of lymphocytes in patients with breast cancer receiving radiotherapy after breast-conserving surgery: A post hoc analysis of a phase III randomized trial. Radiother Oncol 2024; 199:110390. [PMID: 38897316 DOI: 10.1016/j.radonc.2024.110390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/11/2024] [Accepted: 06/16/2024] [Indexed: 06/21/2024]
Abstract
PURPOSE To evaluate the prognostic value of peripheral lymphocyte count (PLC) in the breast cancer patients after breast-conserving surgery (BCS) with radiotherapy (RT). METHODS AND MATERIALS This post hoc analysis was performed using data of 628 patients from a phase III, randomized controlled trial comparing hypofractionated RT (HFRT) with conventional fractionated RT (CFRT) after BCS. PLCs were obtained before, during, and after RT until the 1-year follow-up. The optimal cut-off PLCs were determined using the maxstat package in R. Survival rates were estimated using the Kaplan-Meier method and compared with the log-rank test. RESULTS A total of 275 (46.1 %) patients developed lymphopenia during RT, among them, 17 (2.8 %) had grade 3 lymphopenia and no one developed grade 4 lymphopenia. With a median follow-up of 110.8 months, patients with pre-RT PLCs of < 1.77 × 109/L had a significantly lower 10-year breast cancer-specific survival (BCSS) rate (P = 0.013) and overall survival (OS) rate (P = 0.026). Patients with a nadir PLC of < 1.35 × 109/L had a significantly poorer 10-year OS rate (P = 0.048). Multivariate analysis showed that a pre-RT PLC of < 1.77 × 109/L was an independent factor influencing BCSS and OS, while the effect of the nadir PLC did not remain significant. Neither PLC nor lymphopenia recovery at post-RT 1, 3, and 6 months and 1 year was associated with survival. CONCLUSIONS Radiation-induced lymphopenia in patients with breast cancer after BCS tends to be mild. The lower pre-RT PLC predicted poorer survival.
Collapse
Affiliation(s)
- Yu-Chun Song
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Si-Ye Chen
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xu-Ran Zhao
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hao Jing
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hui Fang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yu Tang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shang-Ying Hu
- Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yong-Wen Song
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jing Jin
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yue-Ping Liu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shu-Nan Qi
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Guang-Yi Sun
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qiu-Zi Zhong
- Department of Radiation Oncology, Beijing Hospital, Ministry of Health, Beijing, China
| | - Xiang-Hui Du
- Department of Radiation Therapy, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, China
| | - Juan Liu
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Ye-Xiong Li
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Shu-Lian Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
34
|
Holtorf C, Mengoni M, Tüting T, Wienke A, Borggrefe J, Surov A, Alter M. [Sarcopenia and immune-related toxicity in patients with malignant melanoma undergoing immune checkpoint inhibition]. DERMATOLOGIE (HEIDELBERG, GERMANY) 2024; 75:798-803. [PMID: 39143426 PMCID: PMC11445294 DOI: 10.1007/s00105-024-05405-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/18/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND AND OBJECTIVES Sarcopenia is the generalized loss of muscle strength, mass, and function. The aim was to investigate whether pretherapeutic sarcopenia, as determined by the psoas muscles, affects therapy-mediated toxicity in patients with malignant melanoma undergoing immunotherapy. PATIENTS AND METHODS Measurement of psoas musculature was performed pretherapeutically using computed tomography at the level of the third lumbar vertebra in the axial plane in 75 patients between January 2011 and December 2020. Sarcopenia was defined using the psoas muscle index (PMI). Immune-related toxicity was retrospectively assessed. RESULTS Treatment-related toxicity was recorded in 33 of the 75 patients (44%). Of these, 16 patients (36.2%) experienced dose-limiting severe events (DLT). Pretherapeutic sarcopenia was identified in 25 patients (33.3%). Comparative analysis showed that the patients with a DLT had lower PMI values compared with the patient group without a DLT (4.65 ± 1.33 vs. 5.79 ± 1.67 cm2m-2, p = 0.015) (odds ratio = 0.60, 95% confidence interval 0.40-0.92, p = 0.02). CONCLUSIONS Pretherapeutic sarcopenia measured based on the psoas muscle is not a significant predictor of immune-mediated toxicity in patients with malignant melanoma treated with immune checkpoint inhibitors. Patients with DLT have lower values for the psoas muscle parameters PMI compared to the group of patients without DLT.
Collapse
Affiliation(s)
- Christian Holtorf
- Hautarztpraxis Dr. med. Anke Raschke, Magdeburg, Deutschland
- Universitätsklinik für Dermatologie und Venerologie, Universitätsklinikum Magdeburg, Magdeburg, Deutschland
| | - Miriam Mengoni
- Universitätsklinik für Dermatologie und Venerologie, Universitätsklinikum Magdeburg, Magdeburg, Deutschland
| | - Thomas Tüting
- Universitätsklinik für Dermatologie und Venerologie, Universitätsklinikum Magdeburg, Magdeburg, Deutschland
| | - Andreas Wienke
- Institut für Medizinische Epidemiologie, Biometrie und Informatik, Martin-Luther-Universität, Halle-Wittenberg, Deutschland
| | - Jan Borggrefe
- Universitätsinstitut für Radiologie, Neuroradiologie und Nuklearmedizin, Johannes-Wesling Klinikum, Ruhr-Universität Bochum, Minden, Deutschland
| | - Alexey Surov
- Universitätsinstitut für Radiologie, Neuroradiologie und Nuklearmedizin, Johannes-Wesling Klinikum, Ruhr-Universität Bochum, Minden, Deutschland
| | - Mareike Alter
- Universitätsklinik für Dermatologie und Venerologie, Ruhr-Universität Bochum, Campus Minden, Johannes-Wesling-Klinikum Minden, Hans-Nolte-Str. 1, 32429, Minden, Deutschland.
| |
Collapse
|
35
|
Surov A, Wienke A, Borggrefe J, Hinnerichs M, Seidensticker R, Öcal O, Schütte K, Zech CJ, Loewe C, van Delden O, Vandecaveye V, Verslype C, Gebauer B, Sengel C, Bargellini I, Iezzi R, Malfertheiner P, Berg T, Klümpen HJ, Benckert J, Gasbarrini A, Amthauer H, Sangro B, Ricke J, Seidensticker M. Skeletal muscle quality predicts overall survival in advanced liver hepatocellular carcinoma treated with SIRT and sorafenib: A subanalysis of the SORAMIC trial. United European Gastroenterol J 2024; 12:1016-1027. [PMID: 39007783 PMCID: PMC11485303 DOI: 10.1002/ueg2.12627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/18/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND AND AIMS Our purpose was to assess the impact of muscle quality on overall survival (OS) in patients with advanced HCC. METHODS This is a subanalysis of the SORAMIC trial. Overall, 363 patients were included. The SIRT/Sorafenib treatment group comprised 182 patients and the sorafenib group 181 patients. Myosteatosis was defined as skeletal muscle density (SMD) < 41 HU for patients with a body mass index up to 24.9 kg/m2 and <33 HU for patients with a body mass index ≥25 kg/m2. Albumin-gauge score was calculated as follows: serum albumin (g/dL) × SMD (HU). To assess the impact of muscle quality on clinical variables and OS, a Cox regression model was used. Hazard ratios are presented together with 95 % confidence intervals (95 % CI). Kaplan-Meier curves were used for survival analysis. RESULTS In the SIRT/sorafenib cohort, low albumin-gauge score was an independent predictor of worse OS, HR = 1.74, CI 95% (1.16-2.62), p = 0.01. In the sorafenib cohort, muscle quality parameters did not predict OS. In alcohol-induced HCC (n = 129), myosteatosis independently predicted OS, HR = 1.85, CI 95% (1.10; 3.12), p = 0.02. In viral-induced HCC (n = 99), parameters of muscle quality did not predict OS. In patients with NASH/Non-alcoholic fatty liver disease (NAFLD) induced HCC, albumin-gauge score was a strong independent predictor of worse OS in the subgroup undergoing combined treatment with SIRT and sorafenib, HR = 9.86, CI 95% (1.12; 86.5), p = 0.04. CONCLUSIONS Myosteatosis predicts independently worse OS in patients with alcohol-induced HCC undergoing combined treatment with SIRT and sorafenib. In patients with NASH/NAFLD induced HCC undergoing treatment with SIRT and sorafenib, albumin-gauge score predicts independently worse OS. IMPACT AND IMPLICATIONS Associations between parameters of muscle quality and OS are different in accordance to the treatment strategy and etiology of HCC. These findings highlight the prognostic potential of skeletal muscle quality in patients with advanced HCC.
Collapse
Affiliation(s)
- Alexey Surov
- Department of RadiologyNeuroradiology and Nuclear MedicineJohannes Wesling University HospitalRuhr University BochumBochumGermany
| | - Andreas Wienke
- Institute of Medical EpidemiologyBiometry and InformaticsUniversity of HalleHalleGermany
| | - Jan Borggrefe
- Department of RadiologyNeuroradiology and Nuclear MedicineJohannes Wesling University HospitalRuhr University BochumBochumGermany
| | - Mattes Hinnerichs
- University Clinic for Radiology and Nuclear MedicineUniversity Hospital MagdeburgMagdeburgGermany
| | | | - Osman Öcal
- Department of RadiologyUniversity HospitalLMU MunichMunichGermany
| | - Kerstin Schütte
- Department of Internal Medicine and GastroenterologyNiels‐Stensen‐Kliniken MarienhospitalOsnabrückGermany
- Klinik für GastroenterologieHepatologie und EndokrinologieMedizinische Hochschule Hannover (MHH)HannoverDeutschland
| | - Christoph J. Zech
- Radiology and Nuclear MedicineUniversity Hospital BaselUniversity of BaselBaselSwitzerland
| | - Christian Loewe
- Section of Cardiovascular and Interventional RadiologyDepartment of Bioimaging and Image‐Guided TherapyMedical University of ViennaViennaAustria
| | - Otto van Delden
- Department of Radiology and Nuclear MedicineAcademic University Medical CentersAmsterdamThe Netherlands
| | | | - Chris Verslype
- Department of Digestive OncologyUniversity Hospitals LeuvenLeuvenBelgium
| | - Bernhard Gebauer
- Department of RadiologyCharité – University Medicine BerlinBerlinGermany
| | - Christian Sengel
- Radiology DepartmentGrenoble University HospitalLa TroncheFrance
| | - Irene Bargellini
- Department of Vascular and Interventional RadiologyUniversity Hospital of PisaPisaItaly
| | - Roberto Iezzi
- Dipartimento di Diagnostica per ImmaginiFondazione Policlinico Universitario A. Gemelli IRCCSUOC di RadiologiaRadioterapia Oncologica ed EmatologiaRomaItalia
| | | | - Thomas Berg
- Klinik und Poliklinik für GastroenterologieSektion HepatologieUniversitätsklinikum LeipzigLeipzigGermany
| | - Heinz J. Klümpen
- Department of Medical OncologyAmsterdam University Medical CentersAmsterdamthe Netherlands
| | - Julia Benckert
- Department of Hepatology and GastroenterologyCharité – Universitätsmedizin BerlinCampus Virchow KlinikumBerlinGermany
| | - Antonio Gasbarrini
- Fondazione Policlinico Universitario Gemelli IRCCSUniversita’ Cattolica del Sacro CuoreRomaItaly
| | - Holger Amthauer
- Department of Nuclear MedicineCharité – Universitätsmedizin BerlinBerlinGermany
| | - Bruno Sangro
- Liver UnitClínica Universidad de Navarra and CIBEREHDPamplonaSpain
| | - Jens Ricke
- Department of RadiologyUniversity HospitalLMU MunichMunichGermany
| | | |
Collapse
|
36
|
Wang W, He S, Zhang W, Zhang H, DeStefano VM, Wada M, Pinz K, Deener G, Shah D, Hagag N, Wang M, Hong M, Zeng R, Lan T, Ma Y, Li F, Liang Y, Guo Z, Zou C, Wang M, Ding L, Ma Y, Yuan Y. BCMA-CD19 compound CAR T cells for systemic lupus erythematosus: a phase 1 open-label clinical trial. Ann Rheum Dis 2024; 83:1304-1314. [PMID: 38777376 DOI: 10.1136/ard-2024-225785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVES This study aims to evaluate the safety and efficacy of BCMA-CD19 compound chimeric antigen receptor T cells (cCAR) to dual reset the humoral and B cell immune system in patients with systemic lupus erythematosus (SLE) with lupus nephritis (LN). METHODS This is a single-arm open-label multicentre phase 1 study of BCMA and CD19-directed cCAR in patients suffering from SLE/LN with autoantibodies produced by B cells and plasma/long-lived plasma cells. In this clinical trial, we sequentially assigned biopsy-confirmed (classes III-V) LN patients to receive 3×106 cCAR cells/kg postcessation of all SLE medications and conditioning. The primary endpoint of safety and toxicity was assessed. Complete immune reset was indicated by B cell receptor (BCR) deep sequencing and flow cytometry analysis. Patient 11 (P11) had insufficient lymphocyte counts and was underdosed as compassionate use. RESULTS P1 and P2 achieved symptom and medication-free remission (MFR) from SLE and complete remission from lymphoma. P3-P13 (excluding P11) received an initial dose of 3×106 cCAR cells /kg and were negative for all autoantibodies, including those derived from long-lived plasma cells, 3 months post-cCAR and the complement returned to normal levels. These patients achieved symptom and MFR with post-cCAR follow-up to 46 months. Complete recovery of B cells was seen in 2-6 months post-cCAR. Mean SLE Disease Activity Index 2000 reduced from 10.6 (baseline) to 2.7 (3 months), and renal function significantly improved in 10 LN patients ≤90 days post-cCAR. cCAR T therapy was well tolerant with mild cytokine-release syndrome. CONCLUSIONS Data suggest that cCAR therapy was safe and effective in inducing MFR and depleting disease-causing autoantibodies in patients with SLE.
Collapse
Affiliation(s)
- Weijia Wang
- Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Shanzhi He
- Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Wenli Zhang
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Hongyu Zhang
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | | | - Masayuki Wada
- iCell Gene Therapeutics Inc, New York, New York, USA
| | - Kevin Pinz
- iCell Gene Therapeutics Inc, New York, New York, USA
| | - Greg Deener
- iCell Gene Therapeutics Inc, New York, New York, USA
| | - Darshi Shah
- iCell Gene Therapeutics Inc, New York, New York, USA
| | - Nabil Hagag
- iCell Gene Therapeutics Inc, New York, New York, USA
| | - Min Wang
- Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Ming Hong
- Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Ronghao Zeng
- Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Ting Lan
- Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Yu Ma
- CAR Bio Therapeutics Ltd, zhongshan, China
| | - Fugui Li
- Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Yingwen Liang
- Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Zhencong Guo
- Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Chanjuan Zou
- Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Mingxia Wang
- Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Ling Ding
- Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Yupo Ma
- iCell Gene Therapeutics Inc, New York, New York, USA
| | - Yong Yuan
- Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| |
Collapse
|
37
|
Praktiknjo M, Pena Solano AS, Sadeghlar F, Welchowski T, Schmid M, Möhring C, Zhou T, Mahn R, Monin MB, Meyer C, Feldmann G, Brossart P, van Beekum C, Semaan A, Matthaei H, Manekeller S, Sprinkart AM, Nowak S, Luetkens J, Kalff JC, Strassburg CP, González-Carmona MA. The impact of lenvatinib on sarcopenia in patients with advanced unresectable hepatocellular carcinoma. Sci Rep 2024; 14:22146. [PMID: 39333610 PMCID: PMC11437060 DOI: 10.1038/s41598-024-66766-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/03/2024] [Indexed: 09/29/2024] Open
Abstract
Lenvatinib is a multiple receptor tyrosine kinase inhibitor (TKI) approved for first-line treatment of patients with unresectable hepatocellular carcinoma (HCC). TKI are suspected of exacerbating muscle loss in patients with cancer. In this study, we analyze the role of muscle loss in patients with advanced HCC treated with lenvatinib. This is a retrospective analysis of a real-life cohort of 25 patients with advanced HCC who were treated with lenvatinib from 2018 to March 2021 in Germany. Patients were stratified for loss of skeletal muscle area during the first three months of lenvatinib therapy. Overall survival (OS), progression-free survival (PFS) and toxicity were analyzed for all patients, especially regarding loss of muscle before and during the first three months of therapy with lenvatinib. Three months after beginning of therapy with lenvatinib, a significant reduction of muscle mass was observed in 60% of patients (p = 0.035). Despite increase of loss of skeletal muscle, patients benefitted from lenvatinib in our cohort of patients in terms of OS and PFS and did not experience increased toxicity. Furthermore, muscle loss was not a negative predictor of survival in the univariate analysis (p = 0.675). Patients with advanced hepatocellular carcinoma experience muscle loss with lenvatinib therapy. However, despite progressive muscle loss, patients benefit from a therapy with lenvatinib in terms of OS and PFS without increased toxicity. However, assessment and prophylaxis of skeletal muscle status should be recommended during a therapy with lenvatinib.
Collapse
Affiliation(s)
- Michael Praktiknjo
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
- Department of Internal Medicine B, University Hospital Münster, Münster, Germany
| | - Ana S Pena Solano
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Farsaneh Sadeghlar
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Thomas Welchowski
- Department of Medical Biometry, Informatics and Epidemiology (IMBIE), University Hospital Bonn, Bonn, Germany
| | - Matthias Schmid
- Department of Medical Biometry, Informatics and Epidemiology (IMBIE), University Hospital Bonn, Bonn, Germany
| | - Christian Möhring
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Taotao Zhou
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Robert Mahn
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Malte B Monin
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Carsten Meyer
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Bonn, Germany
| | - Georg Feldmann
- Department of Internal Medicine III, University Hospital Bonn, Bonn, Germany
| | - Peter Brossart
- Department of Internal Medicine III, University Hospital Bonn, Bonn, Germany
| | - Cornelius van Beekum
- Department of Visceral Surgery, University Hospital Bonn, Bonn, Germany
- Department of General- Abdominal and Transplant Surgery, Hanover Medical School, Hannover, Germany
| | - Alexander Semaan
- Department of Visceral Surgery, University Hospital Bonn, Bonn, Germany
| | - Hanno Matthaei
- Department of Visceral Surgery, University Hospital Bonn, Bonn, Germany
| | | | - Alois M Sprinkart
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Bonn, Germany
| | - Sebastian Nowak
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Bonn, Germany
| | - Julian Luetkens
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Bonn, Germany
| | - Jörg C Kalff
- Department of Visceral Surgery, University Hospital Bonn, Bonn, Germany
| | | | | |
Collapse
|
38
|
Xu J, Gao H, Azhar MS, Xu H, Chen S, Li M, Ni X, Yan T, Zhou H, Long Q, Yi W. Interleukin signaling in the regulation of natural killer cells biology in breast cancer. Front Immunol 2024; 15:1449441. [PMID: 39380989 PMCID: PMC11459090 DOI: 10.3389/fimmu.2024.1449441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 09/09/2024] [Indexed: 10/10/2024] Open
Abstract
In the field of breast cancer treatment, the immunotherapy involving natural killer (NK) cells is increasingly highlighting its distinct potential and significance. Members of the interleukin (IL) family play pivotal regulatory roles in the growth, differentiation, survival, and apoptosis of NK cells, and are central to their anti-tumor activity. These cytokines enhance the ability of NK cells to recognize and eliminate tumor cells by binding to specific receptors and activating downstream signaling pathways. Furthermore, interleukins do not function in isolation; the synergistic or antagonistic interactions between different interleukins can drive NK cells toward various functional pathways, ultimately leading to diverse outcomes for breast cancer patients. This paper reviews the intricate relationship between NK cells and interleukins, particularly within the breast cancer tumor microenvironment. Additionally, we summarize the latest clinical studies and advancements in NK cell therapy for breast cancer, along with the potential applications of interleukin signaling in these therapies. In conclusion, this article underscores the critical role of NK cells and interleukin signaling in breast cancer treatment, providing valuable insights and a significant reference for future research and clinical practice.
Collapse
Affiliation(s)
- Jiachi Xu
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China
| | - Hongyu Gao
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China
| | - Muhammad Salman Azhar
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haifan Xu
- Breast and Thyroid Surgery, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Siyuan Chen
- Breast and Thyroid Surgery, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Mingcan Li
- Breast and Thyroid Surgery, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Xinxi Ni
- Breast and Thyroid Surgery, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Ting Yan
- Breast and Thyroid Surgery, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Hui Zhou
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qian Long
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China
| | - Wenjun Yi
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China
| |
Collapse
|
39
|
Leifheit ME, Johnson G, Kuzel TM, Schneider JR, Barker E, Yun HD, Ustun C, Goldufsky JW, Gupta K, Marzo AL. Enhancing Therapeutic Efficacy of FLT3 Inhibitors with Combination Therapy for Treatment of Acute Myeloid Leukemia. Int J Mol Sci 2024; 25:9448. [PMID: 39273395 PMCID: PMC11394928 DOI: 10.3390/ijms25179448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
FMS-like tyrosine kinase 3 (FLT3) mutations are genetic changes found in approximately thirty percent of patients with acute myeloid leukemia (AML). FLT3 mutations in AML represent a challenging clinical scenario characterized by a high rate of relapse, even after allogeneic hematopoietic stem cell transplantation (allo-HSCT). The advent of FLT3 tyrosine kinase inhibitors (TKIs), such as midostaurin and gilteritinib, has shown promise in achieving complete remission. However, a substantial proportion of patients still experience relapse following TKI treatment, necessitating innovative therapeutic strategies. This review critically addresses the current landscape of TKI treatments for FLT3+ AML, with a particular focus on gilteritinib. Gilteritinib, a highly selective FLT3 inhibitor, has demonstrated efficacy in targeting the mutant FLT3 receptor, thereby inhibiting aberrant signaling pathways that drive leukemic proliferation. However, monotherapy with TKIs may not be sufficient to eradicate AML blasts. Specifically, we provide evidence for integrating gilteritinib with mammalian targets of rapamycin (mTOR) inhibitors and interleukin-15 (IL-15) complexes. The combination of gilteritinib, mTOR inhibitors, and IL-15 complexes presents a compelling strategy to enhance the eradication of AML blasts and enhance NK cell killing, offering a potential for improved patient outcomes.
Collapse
Affiliation(s)
- Malia E Leifheit
- Department of Internal Medicine, Division of Hematology, and Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
| | - Gunnar Johnson
- Department of Internal Medicine, Division of Hematology, and Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
| | - Timothy M Kuzel
- Department of Internal Medicine, Division of Hematology, and Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
| | - Jeffrey R Schneider
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
| | - Edward Barker
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
| | - Hyun D Yun
- Hematology, Oncology, Veterans Affairs Long Beach Healthcare System, Long Beach, CA 90822, USA
- Department of Medicine, Division of Hematology, Oncology, School of Medicine, University of California, Irvine, CA 92617, USA
| | - Celalettin Ustun
- Department of Internal Medicine, Division of Hematology, and Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
| | - Josef W Goldufsky
- Department of Internal Medicine, Division of Hematology, and Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
| | - Kajal Gupta
- Department of Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Amanda L Marzo
- Department of Internal Medicine, Division of Hematology, and Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
40
|
Dejardin D, Kraxner A, Schindler E, Städler N, Wolbers M. An overview of statistical methods for biomarkers relevant to early clinical development of cancer immunotherapies. Front Immunol 2024; 15:1351584. [PMID: 39234243 PMCID: PMC11371698 DOI: 10.3389/fimmu.2024.1351584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 07/29/2024] [Indexed: 09/06/2024] Open
Abstract
Over the last decade, a new paradigm for cancer therapies has emerged which leverages the immune system to act against the tumor. The novel mechanism of action of these immunotherapies has also introduced new challenges to drug development. Biomarkers play a key role in several areas of early clinical development of immunotherapies including the demonstration of mechanism of action, dose finding and dose optimization, mitigation and prevention of adverse reactions, and patient enrichment and indication prioritization. We discuss statistical principles and methods for establishing the prognostic, predictive aspect of a (set of) biomarker and for linking the change in biomarkers to clinical efficacy in the context of early development studies. The methods discussed are meant to avoid bias and produce robust and reproducible conclusions. This review is targeted to drug developers and data scientists interested in the strategic usage and analysis of biomarkers in the context of immunotherapies.
Collapse
Affiliation(s)
- David Dejardin
- Data Sciences, Product Development, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Anton Kraxner
- Roche Pharma Research and Early Development Oncology, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Emilie Schindler
- Roche Pharma Research and Early Development Oncology, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Nicolas Städler
- Roche Pharma Research and Early Development Oncology, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Marcel Wolbers
- Data Sciences, Product Development, F. Hoffmann-La Roche AG, Basel, Switzerland
| |
Collapse
|
41
|
Wittling MC, Cole AC, Brammer B, Diatikar KG, Schmitt NC, Paulos CM. Strategies for Improving CAR T Cell Persistence in Solid Tumors. Cancers (Basel) 2024; 16:2858. [PMID: 39199630 PMCID: PMC11352972 DOI: 10.3390/cancers16162858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024] Open
Abstract
CAR T cells require optimization to be effective in patients with solid tumors. There are many barriers affecting their ability to succeed. One barrier is persistence, as to achieve an optimal antitumor response, infused CAR T cells must engraft and persist. This singular variable is impacted by a multitude of factors-the CAR T cell design, lymphodepletion regimen used, expansion method to generate the T cell product, and more. Additionally, external agents can be utilized to augment CAR T cells, such as the addition of novel cytokines, pharmaceutical drugs that bolster memory formation, or other agents during either the ex vivo expansion process or after CAR T cell infusion to support them in the oppressive tumor microenvironment. This review highlights many strategies being used to optimize T cell persistence as well as future directions for improving the persistence of infused cells.
Collapse
Affiliation(s)
- Megen C. Wittling
- Department of Surgery/Oncology, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322, USA
- School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Anna C. Cole
- Department of Surgery/Oncology, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322, USA
| | - Brianna Brammer
- School of Medicine, Emory University, Atlanta, GA 30322, USA
- Department of Otolaryngology, Emory University, Atlanta, GA 30322, USA
| | - Kailey G. Diatikar
- Department of Surgery/Oncology, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322, USA
| | - Nicole C. Schmitt
- Department of Otolaryngology, Emory University, Atlanta, GA 30322, USA
| | - Chrystal M. Paulos
- Department of Surgery/Oncology, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
42
|
Brown JC, Spielmann G, Yang S, Compton SLE, Jones LW, Irwin ML, Ligibel JA, Meyerhardt JA. Effects of exercise or metformin on myokine concentrations in patients with breast and colorectal cancer: A phase II multi-centre factorial randomized trial. J Cachexia Sarcopenia Muscle 2024; 15:1520-1527. [PMID: 38887915 PMCID: PMC11294014 DOI: 10.1002/jcsm.13509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 05/07/2024] [Accepted: 05/16/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Physical activity and metformin pharmacotherapy are associated with improved clinical outcomes in breast and colorectal cancer survivors. Myokines are cytokines secreted from skeletal muscle that may mediate these associations. METHODS This hypothesis-generating analysis used biospecimens collected from a multi-centre 2 × 2 factorial randomized design of 116 patients with stage I-III breast and colorectal cancer who were randomized to 12 weeks of (1) aerobic exercise (moderate intensity titrated to 220 min/week); (2) metformin (850 mg daily for 2 weeks and then titrated to 850 mg twice per day); (3) aerobic exercise and metformin; or (4) control. Fourteen myokines were quantified using a multiplex panel. Myokine concentrations were log-transformed, and main effects analyses were conducted using linear mixed-effects regression models. The type I error rate was controlled with the Holm sequential testing procedure. RESULTS Randomization to exercise increased leukaemia inhibitory factor (1.26 pg/mL, 95% confidence interval [CI]: 0.69, 1.84; adjusted P = 0.001) and interleukin-15 (2.23 pg/mL, 95% CI: 0.87, 3.60; adjusted P = 0.013) compared with randomization to no exercise. Randomization to metformin decreased apelin (-2.69 pg/mL, 95% CI: -4.31, -1.07; adjusted P = 0.014) and interleukin-15 (-1.74 pg/mL, 95% CI: -2.79, -0.69; adjusted P = 0.013) compared with randomization to no metformin. Metformin decreased myostatin, irisin, oncostatin M, fibroblast growth factor 21 and osteocrin; however, these changes were not statistically significant after correction for multiple comparisons. CONCLUSIONS This pilot study demonstrates that randomization to exercise and metformin elicit unique effects on myokine concentrations in cancer patients. This hypothesis-generating observation warrants further basic, translational and clinical investigation and replication.
Collapse
Affiliation(s)
- Justin C. Brown
- Pennington Biomedical Research CenterBaton RougeLAUSA
- LSU Health Sciences Center New Orleans School of MedicineNew OrleansLAUSA
- Stanley S. Scott Cancer CenterLouisiana State University Health Sciences CenterNew OrleansLAUSA
| | | | | | | | - Lee W. Jones
- Memorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | | | | | | |
Collapse
|
43
|
Luo Z, Zhu J, Xu R, Wan R, He Y, Chen Y, Wang Q, Chen S, Chen S. Exercise-downregulated CD300E acted as a negative prognostic implication and tumor-promoted role in pan-cancer. Front Immunol 2024; 15:1437068. [PMID: 39144140 PMCID: PMC11321962 DOI: 10.3389/fimmu.2024.1437068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/12/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Breast cancer ranks as one of the most prevalent malignancies among women globally, with increasing incidence rates. Physical activity, particularly exercise, has emerged as a potentially significant modifier of cancer prognosis, influencing tumor biology and patient outcomes. METHODS Using a murine breast cancer model, we established a control and an exercise group, where the latter was subjected to 21 days of voluntary running. RNA Sequencing, bioinformatics analysis, pan-cancer analysis, and cell experiments were performed to validate the underlying mechanisms. RESULTS We observed that exercise significantly reduced tumor size and weight, without notable changes in body weight, suggesting that physical activity can modulate tumor dynamics. mRNA sequencing post-exercise revealed substantial downregulation of CD300E in the exercise group, accompanied by alterations in critical pathways such as MicroRNAs in cancers and the Calcium signaling pathway. Expanding our analysis to a broader cancer spectrum, CD300E demonstrated significant expression variability across multiple cancer types, with pronounced upregulation in myeloma, ovarian, lung, and colorectal cancers. This upregulation was correlated with poorer prognostic outcomes, emphasizing CD300E's potential role as a prognostic marker and therapeutic target. Moreover, CD300E expression was associated with cancer cell proliferation and apoptosis. CONCLUSION The study highlights the dual role of exercise in modulating gene expression relevant to tumor growth and the potential of CD300E as a target in cancer therapeutics. Further research is encouraged to explore the mechanisms by which exercise and CD300E influence cancer progression and to develop targeted strategies that could enhance patient outcomes in clinical settings.
Collapse
Affiliation(s)
- Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinguo Zhu
- Department of Orthopaedics, Nantong Tongzhou Hospital of Traditional Chinese Medicine, Tongzhou, Jiangsu, China
| | - Rui Xu
- The First Clinical Medicine College, Nanjing Medical University, Nanjing, China
| | - Renwen Wan
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanwei He
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yisheng Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Qing Wang
- Department of Orthopaedics, Kunshan Hospital of Chinese Medicine, Kunshan, Jiangsu, China
| | - Shuo Chen
- Department of Sports Medicine, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Shiyi Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
44
|
Boersma B, Poinot H, Pommier A. Stimulating the Antitumor Immune Response Using Immunocytokines: A Preclinical and Clinical Overview. Pharmaceutics 2024; 16:974. [PMID: 39204319 PMCID: PMC11357675 DOI: 10.3390/pharmaceutics16080974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/11/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Cytokines are immune modulators which can enhance the immune response and have been proven to be an effective class of immunotherapy. Nevertheless, the clinical use of cytokines in cancer treatment has faced several challenges associated with poor pharmacokinetic properties and the occurrence of adverse effects. Immunocytokines (ICKs) have emerged as a promising approach to overcome the pharmacological limitations observed with cytokines. ICKs are fusion proteins designed to deliver cytokines in the tumor microenvironment by taking advantage of the stability and specificity of immunoglobulin-based scaffolds. Several technological approaches have been developed. This review focuses on ICKs designed with the most impactful cytokines in the cancer field: IL-2, TNFα, IL-10, IL-12, IL-15, IL-21, IFNγ, GM-CSF, and IFNα. An overview of the pharmacological effects of the naked cytokines and ICKs tested for cancer therapy is detailed. A particular emphasis is given on the immunomodulatory effects of ICKs associated with their technological design. In conclusion, this review highlights active ways of development of ICKs. Their already promising results observed in clinical trials are likely to be improved with the advances in targeting technologies such as cytokine/linker engineering and the design of multispecific antibodies with tumor targeting and immunostimulatory functional properties.
Collapse
Affiliation(s)
- Bart Boersma
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland;
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
| | - Hélène Poinot
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland;
- Translational Research Centre in Oncohaematology, University of Geneva, 1211 Geneva, Switzerland
| | - Aurélien Pommier
- UMR1240 Imagerie Moléculaire et Stratégies Théranostiques INSERM, Université Clermont Auvergne, BP 184, F-63005 Clermont-Ferrand, France
| |
Collapse
|
45
|
Gordon MJ, Dubois S, Bryant B, Ng S, Conlon K, Miljkovic MD, Waldmann T, Roschewski M. A phase 1 study of interleukin-15 in combination with avelumab in relapsed or refractory T-cell lymphoma. Leuk Lymphoma 2024; 65:1008-1011. [PMID: 38459780 DOI: 10.1080/10428194.2024.2326847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/29/2024] [Indexed: 03/10/2024]
Affiliation(s)
- Max J Gordon
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Sigrid Dubois
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Bonita Bryant
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Samuel Ng
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Kevin Conlon
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Milos D Miljkovic
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Cartesian therapeutics, Gaithersburg, MD, USA
| | - Thomas Waldmann
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Mark Roschewski
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
46
|
Wang D, Dou L, Sui L, Xue Y, Xu S. Natural killer cells in cancer immunotherapy. MedComm (Beijing) 2024; 5:e626. [PMID: 38882209 PMCID: PMC11179524 DOI: 10.1002/mco2.626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/30/2024] [Accepted: 05/30/2024] [Indexed: 06/18/2024] Open
Abstract
Natural killer (NK) cells, as innate lymphocytes, possess cytotoxic capabilities and engage target cells through a repertoire of activating and inhibitory receptors. Particularly, natural killer group 2, member D (NKG2D) receptor on NK cells recognizes stress-induced ligands-the MHC class I chain-related molecules A and B (MICA/B) presented on tumor cells and is key to trigger the cytolytic response of NK cells. However, tumors have developed sophisticated strategies to evade NK cell surveillance, which lead to failure of tumor immunotherapy. In this paper, we summarized these immune escaping strategies, including the downregulation of ligands for activating receptors, upregulation of ligands for inhibitory receptors, secretion of immunosuppressive compounds, and the development of apoptosis resistance. Then, we focus on recent advancements in NK cell immune therapies, which include engaging activating NK cell receptors, upregulating NKG2D ligand MICA/B expression, blocking inhibitory NK cell receptors, adoptive NK cell therapy, chimeric antigen receptor (CAR)-engineered NK cells (CAR-NK), and NKG2D CAR-T cells, especially several vaccines targeting MICA/B. This review will inspire the research in NK cell biology in tumor and provide significant hope for improving cancer treatment outcomes by harnessing the potent cytotoxic activity of NK cells.
Collapse
Affiliation(s)
- DanRu Wang
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - LingYun Dou
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - LiHao Sui
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - Yiquan Xue
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - Sheng Xu
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
- Shanghai Institute of Stem Cell Research and Clinical Translation Dongfang Hospital Shanghai China
| |
Collapse
|
47
|
Surov A, Wienke A, Gutzmer R, Borggrefe J. Prognostic role of the skeletal musculature in oncology: significance, coherences and clinical implications. ROFO-FORTSCHR RONTG 2024; 196:699-706. [PMID: 38134902 DOI: 10.1055/a-2213-2320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2023]
Affiliation(s)
- Alexey Surov
- Department of Radiology, Neuroradiology and Nuclear Medicine, Johannes Wesling Medical Center, Ruhr University Bochum, Germany
| | - Andreas Wienke
- Institute of Medical Epidemiology, Biostatistics, and Informatics, Martin Luther University Halle Wittenberg, Halle, Germany
| | - Ralf Gutzmer
- Department of Dermatology, Johannes Wesling Medical Center, Ruhr University Bochum, Germany
| | - Jan Borggrefe
- Department of Radiology, Neuroradiology and Nuclear Medicine, Johannes Wesling Medical Center, Ruhr University Bochum, Germany
| |
Collapse
|
48
|
Sánchez-Moreno I, Lasarte-Cia A, Martín-Otal C, Casares N, Navarro F, Gorraiz M, Sarrión P, Hervas-Stubbs S, Jordana L, Rodriguez-Madoz JR, San Miguel J, Prosper F, Lasarte JJ, Lozano T. Tethered IL15-IL15Rα augments antitumor activity of CD19 CAR-T cells but displays long-term toxicity in an immunocompetent lymphoma mouse model. J Immunother Cancer 2024; 12:e008572. [PMID: 38955421 PMCID: PMC11218034 DOI: 10.1136/jitc-2023-008572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Adoptive cell therapy using genetically modified T cells to express chimeric antigen receptors (CAR-T) has shown encouraging results, particularly in certain blood cancers. Nevertheless, over 40% of B cell malignancy patients experience a relapse after CAR-T therapy, likely due to inadequate persistence of the modified T cells in the body. IL15, known for its pro-survival and proliferative properties, has been suggested for incorporation into the fourth generation of CAR-T cells to enhance their persistence. However, the potential systemic toxicity associated with this cytokine warrants further evaluation. METHODS We analyzed the persistence, antitumor efficacy and potential toxicity of anti-mouse CD19 CAR-T cells which express a membrane-bound IL15-IL15Rα chimeric protein (CD19/mbIL15q CAR-T), in BALB/c mice challenged with A20 tumor cells as well as in NSG mice. RESULTS Conventional CD19 CAR-T cells showed low persistence and poor efficacy in BALB/c mice treated with mild lymphodepletion regimens (total body irradiation (TBI) of 1 Gy). CD19/mbIL15q CAR-T exhibits prolonged persistence and enhanced in vivo efficacy, effectively eliminating established A20 B cell lymphoma. However, this CD19/mbIL15q CAR-T displays important long-term toxicities, with marked splenomegaly, weight loss, transaminase elevations, and significant inflammatory findings in some tissues. Mice survival is highly compromised after CD19/mbIL15q CAR-T cell transfer, particularly if a high TBI regimen is applied before CAR-T cell transfer. CONCLUSION Tethered IL15-IL15Rα augments the antitumor activity of CD19 CAR-T cells but displays long-term toxicity in immunocompetent mice. Inducible systems to regulate IL15-IL15Rα expression could be considered to control this toxicity.
Collapse
Affiliation(s)
- Inés Sánchez-Moreno
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, IdISNA, Pamplona, Spain
| | - Aritz Lasarte-Cia
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, IdISNA, Pamplona, Spain
| | - Celia Martín-Otal
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, IdISNA, Pamplona, Spain
| | - Noelia Casares
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, IdISNA, Pamplona, Spain
| | - Flor Navarro
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, IdISNA, Pamplona, Spain
| | - Marta Gorraiz
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, IdISNA, Pamplona, Spain
| | - Patricia Sarrión
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, IdISNA, Pamplona, Spain
| | - Sandra Hervas-Stubbs
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, IdISNA, Pamplona, Spain
- Cancer Center Universidad de Navarra (CCUN), Pamplona, Spain
| | - Lorea Jordana
- Hemato-Oncology Program, Centre for Applied Medical Research (CIMA), University of Navarra, IdiSNA, Pamplona, Spain
| | - Juan Roberto Rodriguez-Madoz
- Cancer Center Universidad de Navarra (CCUN), Pamplona, Spain
- Hemato-Oncology Program, Centre for Applied Medical Research (CIMA), University of Navarra, IdiSNA, Pamplona, Spain
| | - Jesús San Miguel
- Cancer Center Universidad de Navarra (CCUN), Pamplona, Spain
- Hematology and Cell Therapy Department, Clínica Universidad de Navarra, (CUN), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Felipe Prosper
- Cancer Center Universidad de Navarra (CCUN), Pamplona, Spain
- Hemato-Oncology Program, Centre for Applied Medical Research (CIMA), University of Navarra, IdiSNA, Pamplona, Spain
- Hematology and Cell Therapy Department, Clínica Universidad de Navarra, (CUN), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Juan Jose Lasarte
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, IdISNA, Pamplona, Spain
- Cancer Center Universidad de Navarra (CCUN), Pamplona, Spain
| | - Teresa Lozano
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, IdISNA, Pamplona, Spain
- Cancer Center Universidad de Navarra (CCUN), Pamplona, Spain
| |
Collapse
|
49
|
Brummer T, Zeiser R. The role of the MDM2/p53 axis in antitumor immune responses. Blood 2024; 143:2701-2709. [PMID: 37467495 PMCID: PMC11251213 DOI: 10.1182/blood.2023020731] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/26/2023] [Accepted: 07/10/2023] [Indexed: 07/21/2023] Open
Abstract
ABSTRACT Mouse double minute 2 homolog (MDM2) is a negative regulator of the tumor suppressor p53 and is often highly expressed in acute myeloid leukemia (AML) and other solid tumors. Inactivating mutations in TP53, the gene encoding p53, confers an unfavorable prognosis in AML and increases the risk for relapse after allogeneic hematopoietic cell transplantation. We review the concept that manipulation of MDM2 and p53 could enhance immunogenicity of AML and solid tumor cells. Additionally, we discuss the mechanisms by which MDM2 and p53 regulate the expression of major histocompatibility complex class I and II, transcription of double stranded RNA of endogenous retroviruses, responses of interferons, production of interleukin-15, and expression of tumor necrosis factor-related apoptosis-inducing ligand receptor 1 and 2 on malignant cells. The direct effects of MDM2 inhibition or MDM2 deletion in effector T cells are discussed in the context of cancer immunotherapy. The preclinical findings are connected to clinical studies using MDM2 inhibition to enhance antitumor immunity in patients. This review summarizes current evidence supporting the use of MDM2 inhibition to restore p53 as well as the direct effects of MDM2 inhibition on T cells as an emerging concept for combined antitumor immunotherapy against hematological malignancies and beyond.
Collapse
Affiliation(s)
- Tilman Brummer
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Freiburg, Germany
- German Cancer Consortium Partner Site Freiburg and German Cancer Research Center, Heidelberg, Germany
- Signalling Research Centres BIOSS and Centre for Integrative Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Faculty of Medicine, Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Freiburg, Germany
- German Cancer Consortium Partner Site Freiburg and German Cancer Research Center, Heidelberg, Germany
- Signalling Research Centres BIOSS and Centre for Integrative Biological Signaling Studies, University of Freiburg, Freiburg, Germany
- Department of Medicine I (Hematology, Oncology, and Stem Cell Transplantation), Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| |
Collapse
|
50
|
Chu Y, Nayyar G, Tian M, Lee DA, Ozkaynak MF, Ayala-Cuesta J, Klose K, Foley K, Mendelowitz AS, Luo W, Liao Y, Ayello J, Behbehani GK, Riddell S, Cripe T, Cairo MS. Efficiently targeting neuroblastoma with the combination of anti-ROR1 CAR NK cells and N-803 in vitro and in vivo in NB xenografts. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200820. [PMID: 38933492 PMCID: PMC11201149 DOI: 10.1016/j.omton.2024.200820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/02/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024]
Abstract
The prognosis for children with recurrent and/or refractory neuroblastoma (NB) is dismal. The receptor tyrosine kinase-like orphan receptor 1 (ROR1), which is highly expressed on the surface of NB cells, provides a potential target for novel immunotherapeutics. Anti-ROR1 chimeric antigen receptor engineered ex vivo expanded peripheral blood natural killer (anti-ROR1 CAR exPBNK) cells represent this approach. N-803 is an IL-15 superagonist with enhanced biological activity. In this study, we investigated the in vitro and in vivo anti-tumor effects of anti-ROR1 CAR exPBNK cells with or without N-803 against ROR1+ NB models. Compared to mock exPBNK cells, anti-ROR1 CAR exPBNK cells had significantly enhanced cytotoxicity against ROR1+ NB cells, and N-803 further increased cytotoxicity. High-dimensional analysis revealed that N-803 enhanced Stat5 phosphorylation and Ki67 levels in both exPBNK and anti-ROR1 CAR exPBNK cells with or without NB cells. In vivo, anti-ROR1 CAR exPBNK plus N-803 significantly (p < 0.05) enhanced survival in human ROR1+ NB xenografted NSG mice compared to anti-ROR1 CAR exPBNK alone. Our results provide the rationale for further development of anti-ROR1 CAR exPBNK cells plus N-803 as a novel combination immunotherapeutic for patients with recurrent and/or refractory ROR1+ NB.
Collapse
Affiliation(s)
- Yaya Chu
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
| | - Gaurav Nayyar
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
| | - Meijuan Tian
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
| | - Dean A. Lee
- Department of Pediatric Hem/Onc/BMT, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Mehmet F. Ozkaynak
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
| | | | - Kayleigh Klose
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
| | - Keira Foley
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
| | | | - Wen Luo
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
| | - Yanling Liao
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
| | - Janet Ayello
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
| | - Gregory K. Behbehani
- Department of Internal Medicine, Division of Hematology, the Ohio State University; Columbus, OH 43210, USA
| | - Stanley Riddell
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Tim Cripe
- Department of Pediatric Hem/Onc/BMT, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Mitchell S. Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
- Department of Medicine, New York Medical College, Valhalla, NY 10595, USA
- Department of Microbiology, Immunology and Pathology, New York Medical College, Valhalla, NY 10595, USA
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 10595, USA
| |
Collapse
|