1
|
Liang Z, Tian F. Functional nucleic acid-based fluorescence imaging for tumor microenvironment monitoring: A review. Anal Chim Acta 2025; 1350:343794. [PMID: 40155176 DOI: 10.1016/j.aca.2025.343794] [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: 12/11/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND The tumor microenvironment (TME) refers to the complex ecological system surrounding tumor cells, which is intimately associated with regulating tumor cell growth, invasive behavior, and metastatic capacity. Hence, in situ imaging of related bioactivity with resolution in the TME is critical for early cancer detection and accurate diagnosis. In recent years, fluorescence imaging technology has become a widely used tool in TME research due to its non-invasive nature, high spatiotemporal resolution, and capability for real-time monitoring. Among these advancements, signal probes designed based on functional nucleic acids (FNAs) provide a promising and innovative toolkit for targeted imaging analysis of the TME. RESULTS This review provides a comprehensive discussion on the construction of FNA-based biosensors and their advancements in TME monitoring. In this review, we initially provide a systematic summary of the current targeting strategies of FNA-based biosensors for visual monitoring of the TME, focusing on targeting cell surface and extracellular matrix components. Subsequently, we further explore the application of FNA-based biosensors in monitoring the TME. These biosensors have successfully achieved the monitoring of key parameters, bioactive molecules and other tumor markers in the tumor microenvironment due to their excellent molecular recognition ability and high sensitivity. Finally, we discuss some of the challenges currently faced in the field. In response to these challenges, we propose potential research directions and look forward to the future development prospects of this field. SIGNIFICANCE Unlike previous reviews of biosensors based on FNAs for imaging tumor markers in the TME, this work is the first to review how such biosensors can be anchored in the TME. With continued efforts and advancements, we believe an increasing number of FNA-based fluorescence imaging probes will be utilized for TME imaging. This progress will significantly enhance our understanding of disease pathogenesis and progression, thereby offering substantial potential in biosensing and imaging analysis.
Collapse
Affiliation(s)
- Zuoxiang Liang
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, 350122, PR China; Department of Epidemiology and Health Statistics & Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350122, PR China
| | - Fengyu Tian
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, 350122, PR China.
| |
Collapse
|
2
|
White MJV, Raczy MM, Budina E, Yuba E, Solanki A, Shim HN, Zhang ZJ, Gray LT, Cao S, Alpar AT, Hubbell JA. Engineering IL-10 and rapamycin to bind collagen leads to improved anti fibrotic efficacy in lung and kidney fibrosis. Sci Rep 2025; 15:13279. [PMID: 40246931 PMCID: PMC12006466 DOI: 10.1038/s41598-025-94073-3] [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: 02/18/2024] [Accepted: 03/11/2025] [Indexed: 04/19/2025] Open
Abstract
Fibrotic diseases are involved in 45% of deaths in the United States. In particular, fibrosis of the kidney and lung are major public health concerns due to their high prevalence and lack of existing treatment options. Here, we harness the pathophysiological features of fibrotic diseases, namely leaky vasculature and aberrant extracellular matrix (ECM) protein deposition (i.e. collagen), to target an anti-fibrotic biologic and a small molecule drug to disease sites of fibrosis, thus improving the therapeutic potential of both the biologic and small molecule in mouse models of both lung and kidney fibrosis. First, we identify and validate two collagen-targeting drug delivery systems that preferentially accumulate in fibrotic organs: von Willebrand Factor's A3 domain (VWF-A3) and decorin-derived collagen-binding peptide-conjugated micelles (CBP-micelles). We then engineer and recombinantly express novel candidate biologic therapies based on the anti-inflammatory cytokine IL-10: A3-IL-10 and A3-Serum Albumin-IL-10 (A3-SA-IL-10). Simultaneously, we stably encapsulate the potential anti-fibrotic water-insoluble drug, rapamycin, in CBP-micelles. We show that these novel formulations of therapeutics bind to collagen in vitro and that their efficacy in mouse models of lung and kidney fibrosis is improved, compared to free, untargeted drugs. Our results demonstrate that collagen-targeted anti-fibrotic drugs may be next generation therapies of high clinical potential.
Collapse
Affiliation(s)
- Michael J V White
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Michal M Raczy
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Erica Budina
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Eiji Yuba
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Ani Solanki
- Animal Resources Center, University of Chicago, Chicago, IL, 60637, USA
| | - Ha-Na Shim
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Zheng Jenny Zhang
- Comprehensive Transplant Center & Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Laura T Gray
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Shijie Cao
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Aaron T Alpar
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Jeffrey A Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA.
- Committee on Immunology, University of Chicago, Chicago, IL, 60637, USA.
- Committee on Cancer Biology, University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
3
|
Kang S, Mansurov A, Kurtanich T, Chun HR, Slezak AJ, Volpatti LR, Chang K, Wang T, Alpar AT, Refvik KC, Hansen OI, Borjas GJ, Berg BTK, Shim HN, Hultgren KT, Gomes S, Wang Y, Solanki A, Ishihara J, Swartz MA, Hubbell JA. Engineered GM-CSF polarizes protumorigenic tumor-associated macrophages to an antitumorigenic phenotype and potently synergizes with IL-12 immunotherapy. J Immunother Cancer 2024; 12:e009541. [PMID: 39794939 PMCID: PMC11667343 DOI: 10.1136/jitc-2024-009541] [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: 04/23/2024] [Accepted: 11/18/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND The use of immune checkpoint inhibitors (CPIs) has become a dominant regimen in modern cancer therapy, however immune resistance induced by tumor-associated macrophages (TAMs) with immune suppressive and evasion properties limits responses. Therefore, the rational design of immune modulators that can control the immune suppressive properties of TAMs and polarize them, as well as dendritic cells (DCs), toward a more proinflammatory phenotype is a principal objective in cancer immunotherapy. METHODS Here, using a protein engineering approach to enhance cytokine residence in the tumor microenvironment, we examined combined stimulation of the myeloid compartment via tumor stroma-binding granulocyte-macrophage colony-stimulating factor (GM-CSF) to enhance responses in both DCs and T cells via stroma-binding interleukin-12 (IL-12). We evaluated tumor responses at the levels of outcome, cellular responses, and cytokine responses in both the tumors and the tumor-draining lymph nodes. We further investigated the potentiation of DC response to IL-12 by GM-CSF stimulation ex vivo. RESULTS Engineered GM-CSF restored an antitumorigenic tumor myeloid microenvironment otherwise suppressed by TAMs, while engineered IL-12 provided effector signals to T cells, thereby boosting both tumor-resident antitumor macrophage and CD8+ T cell populations. Furthermore, engineered GM-CSF potentiated DC response to IL-12, upregulating DC expression of IL-12 receptor and enhancing their expression of proinflammatory cytokines and chemokines on IL-12 exposure. This resulted in remarkable synergistic efficacy in multiple solid tumor models treated with the dual cytokine combination. The combination therapy also improved the efficacy of CPI in a CPI-resistant genetically-engineered melanoma model and exhibited synergistic antitumor efficacy in a pulmonary metastasis model. CONCLUSION Our strategy provides a rational design for combination immunotherapy targeting both myeloid and lymphoid compartments through complementary mechanisms.
Collapse
Affiliation(s)
- Seounghun Kang
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Aslan Mansurov
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Trevin Kurtanich
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Hye Rin Chun
- Committee on Immunology, The University of Chicago, Chicago, Illinois, USA
| | - Anna J Slezak
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Lisa R Volpatti
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Kevin Chang
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
- Medical Scientist Training Program, The University of Chicago, Chicago, Illinois, USA
| | - Thomas Wang
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Aaron T Alpar
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Kirsten C Refvik
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - O Isabella Hansen
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Gustavo J Borjas
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Brendan T K Berg
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
- Medical Scientist Training Program, The University of Chicago, Chicago, Illinois, USA
| | - Ha-Na Shim
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Kevin T Hultgren
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Suzana Gomes
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Yue Wang
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Ani Solanki
- Animal Resource Center, The University of Chicago, Chicago, Illinois, USA
| | - Jun Ishihara
- Department of Bioengineering, Imperial College London, London, UK
| | - Melody A Swartz
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
- Committee on Immunology, The University of Chicago, Chicago, Illinois, USA
- Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois, USA
- Committee on Cancer Biology, The University of Chicago, Chicago, Illinois, USA
| | - Jeffrey A Hubbell
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
- Committee on Immunology, The University of Chicago, Chicago, Illinois, USA
- Committee on Cancer Biology, The University of Chicago, Chicago, Illinois, USA
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, New York, New York, USA
| |
Collapse
|
4
|
Shi W, Liu N, Lu H. Advancements and challenges in immunocytokines: A new arsenal against cancer. Acta Pharm Sin B 2024; 14:4649-4664. [PMID: 39664443 PMCID: PMC11628837 DOI: 10.1016/j.apsb.2024.07.024] [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: 03/06/2024] [Revised: 05/20/2024] [Accepted: 07/27/2024] [Indexed: 12/13/2024] Open
Abstract
Immunocytokines, employing targeted antibodies to concentrate cytokines at tumor sites, have shown potential advantages such as prolonged cytokine half-lives, mitigated adverse effects, and synergistic antitumor efficacy from both antibody and cytokine components. First, we present an in-depth analysis of the advancements of immunocytokines evaluated in preclinical and clinical applications. Notably, anti-PD-1-based immunocytokines can redirect cytokines to intratumoral CD8+ T cells and reinvigorate them to elicit robust antitumor immune responses. Then, we focus on their molecular structures and action mechanisms, striving to elucidate the correlations between diverse molecular structures and their antitumor efficacy. Moreover, our exploration extends to the realm of novel cytokines, including IL-10, IL-18, and IL-24, unraveling their potential in the construction of immunocytokines. However, safety concerns remain substantial barriers to immunocytokines' development. To address this challenge, we explore potential strategies, such as cytokine engineering and prodrug design, which can foster next-generation immunocytokines development. Overall, this review concentrates on the design of molecular structures in immunocytokines, underscoring the direction and focus of ongoing efforts to improve safety profiles while maximizing therapeutic efficacy.
Collapse
Affiliation(s)
- Wenqiang Shi
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Nan Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Huili Lu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
5
|
Jin SM, Cho JH, Gwak Y, Park SH, Choi K, Choi J, Shin HS, Hong J, Bae Y, Ju J, Shin M, Lim YT. Transformable Gel-to-Nanovaccine Enhances Cancer Immunotherapy via Metronomic-Like Immunomodulation and Collagen-Mediated Paracortex Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409914. [PMID: 39380383 PMCID: PMC11602686 DOI: 10.1002/adma.202409914] [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] [Received: 07/09/2024] [Revised: 09/23/2024] [Indexed: 10/10/2024]
Abstract
The generation of non-exhausted effector T-cells depends on vaccine's spatiotemporal profile, and untimely delivery and low targeting to lymph node (LN) paracortex by standard bolus immunization show limited efficacy. By recapitulating the dynamic processes of acute infection, a bioadhesive immune niche domain (BIND) is developed that facilitates the delivery of timely-activating conjugated nanovaccine (t-CNV) in a metronomic-like manner and increased the accumulation and retention of TANNylated t-CNV (tannic acid coated t-CNV) in LN by specifically binding to collagen in subcapsular sinus where they gradually transformed into TANNylated antigen-adjuvant conjugate by proteolysis, inducing their penetration into paracortex through the collagen-binding in LN conduit and evoking durable antigen-specific CD8+ T-cell responses. The BIND combined with t-CNV, mRNA vaccine, IL-2, and anti-PD-1 antibody also significantly enhanced cancer immunotherapy by the dynamic modulation of immunological landscape of tumor microenvironment. The results provide material design strategy for dynamic immunomodulation that can potentiate non-exhausted T-cell-based immunotherapy.
Collapse
Affiliation(s)
- Seung Mo Jin
- SKKU Advanced Institute of Nanotechnology (SAINT)Department of Nano Engineering, Department of Nano Science and Technology, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKUSungkyunkwan University2066 Seobu‐ro, Jangan‐guSuwonGyeonggi‐do16419Republic of Korea
| | - Ju Hee Cho
- SKKU Advanced Institute of Nanotechnology (SAINT)Department of Nano Engineering, Department of Nano Science and Technology, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKUSungkyunkwan University2066 Seobu‐ro, Jangan‐guSuwonGyeonggi‐do16419Republic of Korea
| | - Yejin Gwak
- SKKU Advanced Institute of Nanotechnology (SAINT)Department of Nano Engineering, Department of Nano Science and Technology, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKUSungkyunkwan University2066 Seobu‐ro, Jangan‐guSuwonGyeonggi‐do16419Republic of Korea
| | - Sei Hyun Park
- SKKU Advanced Institute of Nanotechnology (SAINT)Department of Nano Engineering, Department of Nano Science and Technology, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKUSungkyunkwan University2066 Seobu‐ro, Jangan‐guSuwonGyeonggi‐do16419Republic of Korea
| | - Kyungmin Choi
- Progeneer12 Digital‐ro 31‐gil, Guro‐guSeoul08380Republic of Korea
| | - Jin‐Ho Choi
- SKKU Advanced Institute of Nanotechnology (SAINT)Department of Nano Engineering, Department of Nano Science and Technology, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKUSungkyunkwan University2066 Seobu‐ro, Jangan‐guSuwonGyeonggi‐do16419Republic of Korea
| | - Hong Sik Shin
- SKKU Advanced Institute of Nanotechnology (SAINT)Department of Nano Engineering, Department of Nano Science and Technology, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKUSungkyunkwan University2066 Seobu‐ro, Jangan‐guSuwonGyeonggi‐do16419Republic of Korea
| | - JungHyub Hong
- Department of Biological SciencesScience Research Center (SRC) for Immune Research on Non‐lymphoid Organ (CIRNO)Sungkyunkwan University2066 Seobu‐ro, Jangan‐guSuwonGyeonggi‐do16419Republic of Korea
| | - Yong‐Soo Bae
- Department of Biological SciencesScience Research Center (SRC) for Immune Research on Non‐lymphoid Organ (CIRNO)Sungkyunkwan University2066 Seobu‐ro, Jangan‐guSuwonGyeonggi‐do16419Republic of Korea
| | - Jaewon Ju
- Department of Biomedical EngineeringCenter for Neuroscience Imaging ResearchInstitute for Basic Science (IBS)Department of Intelligent Precision Healthcare ConvergenceSungkyunkwan University2066 Seobu‐ro, Jangan‐guSuwonGyeonggi‐do16419Republic of Korea
| | - Mikyung Shin
- Department of Biomedical EngineeringCenter for Neuroscience Imaging ResearchInstitute for Basic Science (IBS)Department of Intelligent Precision Healthcare ConvergenceSungkyunkwan University2066 Seobu‐ro, Jangan‐guSuwonGyeonggi‐do16419Republic of Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT)Department of Nano Engineering, Department of Nano Science and Technology, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKUSungkyunkwan University2066 Seobu‐ro, Jangan‐guSuwonGyeonggi‐do16419Republic of Korea
| |
Collapse
|
6
|
Stinson JA, Barbosa MMP, Sheen A, Momin N, Fink E, Hampel J, Selting KA, Kamerer RL, Bailey KL, Wittrup KD, Fan TM. Tumor-Localized Interleukin-2 and Interleukin-12 Combine with Radiation Therapy to Safely Potentiate Regression of Advanced Malignant Melanoma in Pet Dogs. Clin Cancer Res 2024; 30:4029-4043. [PMID: 38980919 PMCID: PMC11398984 DOI: 10.1158/1078-0432.ccr-24-0861] [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: 03/15/2024] [Revised: 05/08/2024] [Accepted: 07/03/2024] [Indexed: 07/11/2024]
Abstract
PURPOSE Cytokines IL2 and IL12 exhibit potent anticancer activity but suffer a narrow therapeutic window due to off-tumor immune cell activation. Engineering cytokines with the ability to bind and associate with tumor collagen after intratumoral injection potentiated response without toxicity in mice and was previously safe in pet dogs with sarcoma. Here, we sought to test the efficacy of this approach in dogs with advanced melanoma. PATIENTS AND METHODS This study examined 15 client-owned dogs with histologically or cytologically confirmed malignant melanoma that received a single 9-Gy fraction of radiotherapy, followed by six cycles of combined collagen-anchored IL2 and IL12 therapy every 2 weeks. Cytokine dosing followed a 3 + 3 dose escalation design, with the initial cytokine dose chosen from prior evaluation in canine sarcomas. No exclusion criteria for tumor stage or metastatic burden, age, weight, or neuter status were applied for this trial. RESULTS Median survival regardless of the tumor stage or dose level was 256 days, and 10/13 (76.9%) dogs that completed treatment had CT-measured tumor regression at the treated lesion. In dogs with metastatic disease, 8/13 (61.5%) had partial responses across their combined lesions, which is evidence of locoregional response. Profiling by NanoString of treatment-resistant dogs revealed that B2m loss was predictive of poor response to this therapy. CONCLUSIONS Collectively, these results confirm the ability of locally administered tumor-anchored cytokines to potentiate responses at regional disease sites when combined with radiation. This evidence supports the clinical translation of this approach and highlights the utility of comparative investigation in canine cancers.
Collapse
Affiliation(s)
- Jordan A. Stinson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | | | - Allison Sheen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Noor Momin
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA
| | - Elizabeth Fink
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Jordan Hampel
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Kim A. Selting
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Rebecca L. Kamerer
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL
| | | | - K. Dane Wittrup
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Timothy M. Fan
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL
| |
Collapse
|
7
|
Chen S, Li K, Chen X, Lei S, Lin J, Huang P. Reversibly photoswitchable protein assemblies with collagen affinity for in vivo photoacoustic imaging of tumors. SCIENCE ADVANCES 2024; 10:eadn8274. [PMID: 39213344 PMCID: PMC11364091 DOI: 10.1126/sciadv.adn8274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
Recent advancements in photoacoustic (PA) imaging have leveraged reversibly photoswitchable chromophores, known for their dual absorbance states, to enhance imaging sensitivity through differential techniques. Yet, their deployment in tumor imaging has faced obstacles in achieving targeted delivery with high efficiency and specificity. Addressing this challenge, we introduce innovative protein assemblies, DrBphP-CBD, by genetically fusing a photosensory module from Deinococcus radiodurans bacterial phytochrome (DrBphP) with a collagen-binding domain (CBD). These protein assemblies form sub-100-nanometer structures composed of 24 DrBphP dimers and 12 CBD trimers, presenting 24 protein subunits. Their affinity for collagens, combined with impressive photoswitching contrast, markedly improves PA imaging precision. In various tumor models, intravenous administration of DrBphP-CBD has demonstrated enhanced tumor targeting and retention, augmenting contrast in PA imaging by minimizing background noise. This strategy underscores the clinical potential of DrBphP-CBD as PA contrast agents, propelling photoswitchable chromoproteins to the forefront of precise cancer diagnosis.
Collapse
Affiliation(s)
| | | | - Xin Chen
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Shan Lei
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | | | | |
Collapse
|
8
|
Prakash J, Shaked Y. The Interplay between Extracellular Matrix Remodeling and Cancer Therapeutics. Cancer Discov 2024; 14:1375-1388. [PMID: 39091205 PMCID: PMC11294818 DOI: 10.1158/2159-8290.cd-24-0002] [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: 01/02/2024] [Revised: 04/12/2024] [Accepted: 05/22/2024] [Indexed: 08/04/2024]
Abstract
The extracellular matrix (ECM) is an abundant noncellular component of most solid tumors known to support tumor progression and metastasis. The interplay between the ECM and cancer therapeutics opens up new avenues in understanding cancer biology. While the ECM is known to protect the tumor from anticancer agents by serving as a biomechanical barrier, emerging studies show that various cancer therapies induce ECM remodeling, resulting in therapy resistance and tumor progression. This review discusses critical issues in this field including how the ECM influences treatment outcome, how cancer therapies affect ECM remodeling, and the challenges associated with targeting the ECM. Significance: The intricate relationship between the extracellular matrix (ECM) and cancer therapeutics reveals novel insights into tumor biology and its effective treatment. While the ECM may protect tumors from anti-cancer agents, recent research highlights the paradoxical role of therapy-induced ECM remodeling in promoting treatment resistance and tumor progression. This review explores the key aspects of the interplay between ECM and cancer therapeutics.
Collapse
Affiliation(s)
- Jai Prakash
- Engineered Therapeutics Group, Department of Advanced Organ Bioengineering and Therapeutics, Technical Medical Centre, University of Twente, Enschede, the Netherlands.
| | - Yuval Shaked
- Rappaport Faculty of Medicine, Rappaport-Technion Integrated Cancer Center, Technion – Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
9
|
Mehta NK, Rakhra K, Meetze KA, Li B, Momin N, Chang JY, Wittrup KD, Baeuerle PA, Michaelson JS. CLN-617 Retains IL2 and IL12 in Injected Tumors to Drive Robust and Systemic Immune-Mediated Antitumor Activity. Cancer Immunol Res 2024; 12:1022-1038. [PMID: 38842347 PMCID: PMC11292205 DOI: 10.1158/2326-6066.cir-23-0636] [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: 08/03/2023] [Revised: 02/07/2024] [Accepted: 05/31/2024] [Indexed: 06/07/2024]
Abstract
Despite clinical evidence of antitumor activity, the development of cytokine therapies has been hampered by a narrow therapeutic window and limited response rates. Two cytokines of high interest for clinical development are interleukin 2 (IL2) and interleukin 12 (IL12), which potently synergize to promote the activation and proliferation of T cells and NK cells. However, the only approved human IL2 therapy, Proleukin, is rarely used in the clinic due to systemic toxicities, and no IL12 product has been approved to date due to severe dose-limiting toxicities. Here, we describe CLN-617, a first-in-class therapeutic for intratumoral (IT) injection that co-delivers IL2 and IL12 on a single molecule in a safe and effective manner. CLN-617 is a single-chain fusion protein comprised of IL2, leukocyte-associated immunoglobulin-like receptor 2 (LAIR2), human serum albumin (HSA), and IL12. LAIR2 and HSA function to retain CLN-617 in the treated tumor by binding collagen and increasing molecular weight, respectively. We found that IT administration of a murine surrogate of CLN-617, mCLN-617, eradicated established treated and untreated tumors in syngeneic models, significantly improved response to anti-PD1 checkpoint therapy, and generated a robust abscopal response dependent on cellular immunity and antigen cross-presentation. CLN-617 is being evaluated in a clinical trial in patients with advanced solid tumors (NCT06035744).
Collapse
Affiliation(s)
| | - Kavya Rakhra
- Cullinan Therapeutics, Inc., Cambridge, Massachusetts.
| | | | - Bochong Li
- Cullinan Therapeutics, Inc., Cambridge, Massachusetts.
| | - Noor Momin
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania.
| | | | - K. Dane Wittrup
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.
| | - Patrick A. Baeuerle
- Cullinan Therapeutics, Inc., Cambridge, Massachusetts.
- Institute for Immunology, Ludwig Maximilians University, München, Germany.
| | | |
Collapse
|
10
|
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
|
11
|
Revert-Ros F, Ventura I, Prieto-Ruiz JA, Hernández-Andreu JM, Revert F. The Versatility of Collagen in Pharmacology: Targeting Collagen, Targeting with Collagen. Int J Mol Sci 2024; 25:6523. [PMID: 38928229 PMCID: PMC11203716 DOI: 10.3390/ijms25126523] [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: 05/08/2024] [Revised: 06/01/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
Collagen, a versatile family of proteins with 28 members and 44 genes, is pivotal in maintaining tissue integrity and function. It plays a crucial role in physiological processes like wound healing, hemostasis, and pathological conditions such as fibrosis and cancer. Collagen is a target in these processes. Direct methods for collagen modulation include enzymatic breakdown and molecular binding approaches. For instance, Clostridium histolyticum collagenase is effective in treating localized fibrosis. Polypeptides like collagen-binding domains offer promising avenues for tumor-specific immunotherapy and drug delivery. Indirect targeting of collagen involves regulating cellular processes essential for its synthesis and maturation, such as translation regulation and microRNA activity. Enzymes involved in collagen modification, such as prolyl-hydroxylases or lysyl-oxidases, are also indirect therapeutic targets. From another perspective, collagen is also a natural source of drugs. Enzymatic degradation of collagen generates bioactive fragments known as matrikines and matricryptins, which exhibit diverse pharmacological activities. Overall, collagen-derived peptides present significant therapeutic potential beyond tissue repair, offering various strategies for treating fibrosis, cancer, and genetic disorders. Continued research into specific collagen targeting and the application of collagen and its derivatives may lead to the development of novel treatments for a range of pathological conditions.
Collapse
Affiliation(s)
| | | | | | | | - Fernando Revert
- Mitochondrial and Molecular Medicine Research Group, Facultad de Medicina y Ciencias de la Salud, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain; (F.R.-R.); (I.V.); (J.A.P.-R.); (J.M.H.-A.)
| |
Collapse
|
12
|
Borst R, Meyaard L, Pascoal Ramos MI. Understanding the matrix: collagen modifications in tumors and their implications for immunotherapy. J Transl Med 2024; 22:382. [PMID: 38659022 PMCID: PMC11040975 DOI: 10.1186/s12967-024-05199-3] [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: 12/15/2023] [Accepted: 04/13/2024] [Indexed: 04/26/2024] Open
Abstract
Tumors are highly complex and heterogenous ecosystems where malignant cells interact with healthy cells and the surrounding extracellular matrix (ECM). Solid tumors contain large ECM deposits that can constitute up to 60% of the tumor mass. This supports the survival and growth of cancerous cells and plays a critical role in the response to immune therapy. There is untapped potential in targeting the ECM and cell-ECM interactions to improve existing immune therapy and explore novel therapeutic strategies. The most abundant proteins in the ECM are the collagen family. There are 28 different collagen subtypes that can undergo several post-translational modifications (PTMs), which alter both their structure and functionality. Here, we review current knowledge on tumor collagen composition and the consequences of collagen PTMs affecting receptor binding, cell migration and tumor stiffness. Furthermore, we discuss how these alterations impact tumor immune responses and how collagen could be targeted to treat cancer.
Collapse
Affiliation(s)
- Rowie Borst
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Linde Meyaard
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - M Ines Pascoal Ramos
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
- Oncode Institute, Utrecht, The Netherlands.
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal.
| |
Collapse
|
13
|
Niu L, Jang E, Chin AL, Huo Z, Wang W, Cai W, Tong R. Noncovalently particle-anchored cytokines with prolonged tumor retention safely elicit potent antitumor immunity. SCIENCE ADVANCES 2024; 10:eadk7695. [PMID: 38640236 PMCID: PMC11029804 DOI: 10.1126/sciadv.adk7695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 03/19/2024] [Indexed: 04/21/2024]
Abstract
Preclinical studies have shown that immunostimulatory cytokines elicit antitumor immune responses but their clinical use is limited by severe immune-related adverse events upon systemic administration. Here, we report a facile and versatile strategy for noncovalently anchoring potent Fc-fused cytokine molecules to the surface of size-discrete particles decorated with Fc-binding peptide for local administration. Following intratumoral injection, particle-anchored Fc cytokines exhibit size-dependent intratumoral retention. The 1-micrometer particle prolongs intratumoral retention of Fc cytokine for over a week and has minimal systemic exposure, thereby eliciting antitumor immunity while eliminating systemic toxicity caused by circulating cytokines. In addition, the combination of these particle-anchored cytokines with immune checkpoint blockade antibodies safely promotes tumor regression in various syngeneic tumor models and genetically engineered murine tumor models and elicits systemic antitumor immunity against tumor rechallenge. Our formulation strategy renders a safe and tumor-agnostic approach that uncouples cytokines' immunostimulatory properties from their systemic toxicities for potential clinical application.
Collapse
Affiliation(s)
- Liqian Niu
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, 635 Prices Fork Road, Blacksburg, VA, 24061, USA
| | - Eungyo Jang
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, 635 Prices Fork Road, Blacksburg, VA, 24061, USA
| | - Ai Lin Chin
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, 635 Prices Fork Road, Blacksburg, VA, 24061, USA
| | - Ziyu Huo
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, 635 Prices Fork Road, Blacksburg, VA, 24061, USA
| | - Wenbo Wang
- Department of Materials Science and Engineering, Virginia Polytechnic Institute and State University, 445 Old Turner Street, Blacksburg, VA, 24061, USA
| | - Wenjun Cai
- Department of Materials Science and Engineering, Virginia Polytechnic Institute and State University, 445 Old Turner Street, Blacksburg, VA, 24061, USA
| | - Rong Tong
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, 635 Prices Fork Road, Blacksburg, VA, 24061, USA
| |
Collapse
|
14
|
Zhang X, Yang Q, Zhou S, Li C, Jiang X. Dynamic monitoring of the fibrosis disease by a collagen targeting near infrared probe. Biomater Sci 2024; 12:1924-1931. [PMID: 38437021 DOI: 10.1039/d3bm01926h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
The deposition of the extracellular matrix, especially collagen, and the elevated expression levels of reactive oxygen species, including H2O2, are the main features of fibrosis. Fibrosis can occur in many tissues, such as tumor and liver tissues. The deposition of collagen in the location of lesions not only leads to immunological rejection and supports liver fibrosis and tumor progression, but also provides unique physiological signals with the progression of fibrosis and tumor. However, at present, the detection of fibrosis, especially real time detection, is greatly difficult, making it important to develop noninvasive probes for the dynamic monitoring of fibrosis progression. Herein, we propose a H2O2 responsive macromolecular probe for collagen imaging with high sensitivity and specificity. This probe consists of a collagen-targeting peptide and a H2O2-sensitive and near-infrared (NIR)-emitting macromolecular optical probe, which could effectively bind to collagen both in vitro and in vivo in the region of tumor or fibrotic liver tissues, allowing for high sensitivity and noninvasive visualization of fibrotic tissues and real time monitoring of collagen degradation after anti-fibrotic drug treatment.
Collapse
Affiliation(s)
- Xiaoke Zhang
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Qianwen Yang
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Sensen Zhou
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Cheng Li
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Xiqun Jiang
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
15
|
Wang S, Li Y, Xu C, Dong J, Wei J. An oncolytic vaccinia virus encoding hyaluronidase reshapes the extracellular matrix to enhance cancer chemotherapy and immunotherapy. J Immunother Cancer 2024; 12:e008431. [PMID: 38458640 PMCID: PMC10921532 DOI: 10.1136/jitc-2023-008431] [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: 02/05/2024] [Indexed: 03/10/2024] Open
Abstract
BACKGROUND The redundant extracellular matrix (ECM) within tumor microenvironment (TME) such as hyaluronic acid (HA) often impairs intratumoral dissemination of antitumor drugs. Oncolytic viruses (OVs) are being studied extensively for cancer therapy either alone or in conjunction with chemotherapy and immunotherapy. Here, we designed a novel recombinant vaccinia virus encoding a soluble version of hyaluronidase Hyal1 (OVV-Hyal1) to degrade the HA and investigated its antitumor effects in combination with chemo drugs, polypeptide, immune cells, and antibodies. METHODS We constructed a recombinant oncolytic vaccinia virus encoding the hyaluronidase, and investigated its function in remodeling the ECM of the TME, the antitumor efficacy both in vitro and in several murine solid tumors either alone, or in combination with chemo drugs including doxorubicin and gemcitabine, with polypeptide liraglutide, with immune therapeutics such as PD-L1/PD-1 blockade, CD47 antibody, and with CAR-T cells. RESULTS Compared with control OVV, intratumoral injection of OVV-Hyal1 showed superior antitumor efficacies in a series of mouse subcutaneous tumor models. Moreover, HA degradation by OVV-Hyal1 resulted in increased intratumoral dissemination of chemo drugs, infiltration of T cells, NK cells, macrophages, and activation of CD8+ T cells. When OVV-Hyal1 was combined with some antitumor therapeutics, for example, doxorubicin, gemcitabine, liraglutide, anti-PD-1, anti-CD47 blockade, or CAR-T cells, more profound therapeutic outcomes were obtained. CONCLUSIONS OVV-Hyal1 effectively degrades HA to reshape the TME, therefore overcoming some major hurdles in current cancer therapy, such as limited OVs spread, unfavored dissemination of chemo drugs, polypeptides, antibodies, and insufficient infiltration of effector immune cells. OVV-Hyal1 holds the promise to improve the antitumor outcomes of current cancer therapeutics.
Collapse
Affiliation(s)
- Shibing Wang
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
- Cancer Center, Department of Pathology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuxin Li
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Chuning Xu
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Jie Dong
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Jiwu Wei
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
16
|
Qin Y, Wang G, Chen L, Sun Y, Yang J, Piao Y, Shen Y, Zhou Z. High-Throughput Screening of Surface Engineered Cyanine Nanodots for Active Transport of Therapeutic Antibodies into Solid Tumor. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2302292. [PMID: 37405862 DOI: 10.1002/adma.202302292] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/08/2023] [Accepted: 07/03/2023] [Indexed: 07/07/2023]
Abstract
The successful delivery of therapeutic biomacromolecules into solid tumor holds great challenge due to their high resistance to penetrate through the complex tumor microenvironments. Here, active-transporting nanoparticles are harnessed to efficiently deliver biomacromolecular drugs into solid tumors through cell transcytosis. A series of molecularly precise cyanine 5-cored polylysine G5 dendrimers (Cy5 nanodots) with different peripheral amino acids (G5-AA) is prepared. The capability of these positively charged nanodots to induce cell endocytosis, exocytosis, and transcytosis is evaluated via fluorescence-based high-throughput screen. The optimized nanodots (G5-R) are conjugated with αPD-L1 (a therapeutic monoclonal antibody binding to programmed-death ligand 1) (αPD-L1-G5-R) to demonstrate the nanoparticle-mediated tumor active transport. The αPD-L1-G5-R can greatly enhance the tumor-penetration capability through adsorption-mediated transcytosis (AMT). The effectiveness of αPD-L1-G5-R is tested in treating mice bearing partially resected CT26 tumors, mimicking the local immunotherapy of residual tumors post-surgery in clinic. The αPD-L1-G5-R embedded in fibrin gel can efficiently mediate tumor cell transcytosis, and deliver αPD-L1 throughout the tumor, thereby enhancing immune checkpoint blockade, reducing tumor recurrence, and significantly prolonging the survival time. The active-transporting nanodots are promising platforms for efficient tumor delivery of therapeutic biomacromolecules.
Collapse
Affiliation(s)
- Yating Qin
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311200, China
| | - Guowei Wang
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311200, China
| | - Linying Chen
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yuji Sun
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jiajia Yang
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
17
|
Palmeri JR, Lax BM, Peters JM, Duhamel L, Stinson JA, Santollani L, Lutz EA, Pinney W, Bryson BD, Dane Wittrup K. CD8 + T cell priming that is required for curative intratumorally anchored anti-4-1BB immunotherapy is constrained by Tregs. Nat Commun 2024; 15:1900. [PMID: 38429261 PMCID: PMC10907589 DOI: 10.1038/s41467-024-45625-0] [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: 03/16/2023] [Accepted: 01/30/2024] [Indexed: 03/03/2024] Open
Abstract
Although co-stimulation of T cells with agonist antibodies targeting 4-1BB (CD137) improves antitumor immune responses in preclinical studies, clinical success has been limited by on-target, off-tumor activity. Here, we report the development of a tumor-anchored ɑ4-1BB agonist (ɑ4-1BB-LAIR), which consists of a ɑ4-1BB antibody fused to the collagen-binding protein LAIR. While combination treatment with an antitumor antibody (TA99) shows only modest efficacy, simultaneous depletion of CD4+ T cells boosts cure rates to over 90% of mice. Mechanistically, this synergy depends on ɑCD4 eliminating tumor draining lymph node regulatory T cells, resulting in priming and activation of CD8+ T cells which then infiltrate the tumor microenvironment. The cytotoxic program of these newly primed CD8+ T cells is then supported by the combined effect of TA99 and ɑ4-1BB-LAIR. The combination of TA99 and ɑ4-1BB-LAIR with a clinically approved ɑCTLA-4 antibody known for enhancing T cell priming results in equivalent cure rates, which validates the mechanistic principle, while the addition of ɑCTLA-4 also generates robust immunological memory against secondary tumor rechallenge. Thus, our study establishes the proof of principle for a clinically translatable cancer immunotherapy.
Collapse
Affiliation(s)
- Joseph R Palmeri
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Brianna M Lax
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Joshua M Peters
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Lauren Duhamel
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Jordan A Stinson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Luciano Santollani
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Emi A Lutz
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - William Pinney
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Bryan D Bryson
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - K Dane Wittrup
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA.
| |
Collapse
|
18
|
Stinson JA, Barbosa MMP, Sheen A, Momin N, Fink E, Hampel J, Selting K, Kamerer R, Bailey KL, Wittrup KD, Fan TM. Tumor-localized interleukin-2 and interleukin-12 combine with radiation therapy to safely potentiate regression of advanced malignant melanoma in pet dogs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.12.579965. [PMID: 38405716 PMCID: PMC10888855 DOI: 10.1101/2024.02.12.579965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
The clinical use of interleukin-2 and -12 cytokines against cancer is limited by their narrow therapeutic windows due to on-target, off-tumor activation of immune cells when delivered systemically. Engineering IL-2 and IL-12 to bind to extracellular matrix collagen allows these cytokines to be retained within tumors after intralesional injection, overcoming these clinical safety challenges. While this approach has potentiated responses in syngeneic mouse tumors without toxicity, the complex tumor-immune interactions in human cancers are difficult to recapitulate in mouse models of cancer. This has driven an increased role for comparative oncology clinical trials in companion (pet) dogs with spontaneous cancers that feature analogous tumor and immune biology to human cancers. Here, we report the results from a dose-escalation clinical trial of intratumoral collagen-binding IL-2 and IL-12 cytokines in pet dogs with malignant melanoma, observing encouraging local and regional responses to therapy that may suggest human clinical benefit with this approach.
Collapse
Affiliation(s)
- Jordan A. Stinson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | | | - Allison Sheen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Noor Momin
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA
| | - Elizabeth Fink
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Jordan Hampel
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Kimberly Selting
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Rebecca Kamerer
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL
| | | | - K. Dane Wittrup
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Timothy M. Fan
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL
| |
Collapse
|
19
|
Collado J, Boland L, Ahrendsen JT, Miska J, Lee-Chang C. Understanding the glioblastoma tumor microenvironment: leveraging the extracellular matrix to increase immunotherapy efficacy. Front Immunol 2024; 15:1336476. [PMID: 38380331 PMCID: PMC10876826 DOI: 10.3389/fimmu.2024.1336476] [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: 11/10/2023] [Accepted: 01/24/2024] [Indexed: 02/22/2024] Open
Abstract
Glioblastoma (GBM) accounts for approximately half of all malignant brain tumors, and it remains lethal with a five-year survival of less than 10%. Despite the immense advancements in the field, it has managed to evade even the most promising therapeutics: immunotherapies. The main reason is the highly spatiotemporally heterogeneous and immunosuppressive GBM tumor microenvironment (TME). Accounting for this complex interplay of TME-driven immunosuppression is key to developing effective therapeutics. This review will explore the immunomodulatory role of the extracellular matrix (ECM) by establishing its contribution to the TME as a key mediator of immune responses in GBM. This relationship will help us elucidate therapeutic targets that can be leveraged to develop and deliver more effective immunotherapies.
Collapse
Affiliation(s)
- Jimena Collado
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Lauren Boland
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, United States
| | - Jared T Ahrendsen
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jason Miska
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Lurie Cancer Center, Lou and Jean Malnati Brain Tumor Institute, Chicago, IL, United States
| | - Catalina Lee-Chang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Lurie Cancer Center, Lou and Jean Malnati Brain Tumor Institute, Chicago, IL, United States
| |
Collapse
|
20
|
Shen X, Pan D, Gong Q, Gu Z, Luo K. Enhancing drug penetration in solid tumors via nanomedicine: Evaluation models, strategies and perspectives. Bioact Mater 2024; 32:445-472. [PMID: 37965242 PMCID: PMC10641097 DOI: 10.1016/j.bioactmat.2023.10.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/18/2023] [Accepted: 10/18/2023] [Indexed: 11/16/2023] Open
Abstract
Effective tumor treatment depends on optimizing drug penetration and accumulation in tumor tissue while minimizing systemic toxicity. Nanomedicine has emerged as a key solution that addresses the rapid clearance of free drugs, but achieving deep drug penetration into solid tumors remains elusive. This review discusses various strategies to enhance drug penetration, including manipulation of the tumor microenvironment, exploitation of both external and internal stimuli, pioneering nanocarrier surface engineering, and development of innovative tactics for active tumor penetration. One outstanding strategy is organelle-affinitive transfer, which exploits the unique properties of specific tumor cell organelles and heralds a potentially transformative approach to active transcellular transfer for deep tumor penetration. Rigorous models are essential to evaluate the efficacy of these strategies. The patient-derived xenograft (PDX) model is gaining traction as a bridge between laboratory discovery and clinical application. However, the journey from bench to bedside for nanomedicines is fraught with challenges. Future efforts should prioritize deepening our understanding of nanoparticle-tumor interactions, re-evaluating the EPR effect, and exploring novel nanoparticle transport mechanisms.
Collapse
Affiliation(s)
- Xiaoding Shen
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
| | - Dayi Pan
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, 361021, China
| | - Zhongwei Gu
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
21
|
Tomala J, Cao SD, Spangler JB. Engineering Anticytokine Antibodies for Immune Modulation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:225-234. [PMID: 38166248 DOI: 10.4049/jimmunol.2300467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/28/2023] [Indexed: 01/04/2024]
Abstract
The delicate balance of immune homeostasis is regulated by the interactions between cytokines and their cognate cell surface signaling receptors. There is intensive interest in harnessing cytokines as drugs for diseases such as cancer and autoimmune disorders. However, the multifarious and often contradictory activities of cytokines, coupled with their short serum half-lives, limit clinical performance and result in dangerous toxicities. There is thus growing emphasis on manipulating natural cytokines to enhance their selectivity, safety, and durability through various strategies. One strategy that has gained traction in recent years is the development of anticytokine Abs that not only extend the circulation half-life of cytokines but also specifically bias their immune activities through multilayered molecular mechanisms. Although Abs are notorious for their antagonistic activities, this review focuses on anticytokine Abs that selectively agonize the activity of the target protein. This approach has potential to help realize the clinical promise of cytokine-based therapies.
Collapse
Affiliation(s)
- Jakub Tomala
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University School of Engineering, Baltimore, MD
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Shanelle D Cao
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University School of Engineering, Baltimore, MD
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jamie B Spangler
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University School of Engineering, Baltimore, MD
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
22
|
Elsafy S, Metselaar J, Lammers T. Nanomedicine - Immune System Interactions: Limitations and Opportunities for the Treatment of Cancer. Handb Exp Pharmacol 2024; 284:231-265. [PMID: 37578622 DOI: 10.1007/164_2023_685] [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] [Indexed: 08/15/2023]
Abstract
Nanoparticles interact with immune cells in many different ways. These interactions are crucially important for determining nanoparticles' ability to be used for cancer therapy. Traditionally, strategies such as PEGylation have been employed to reduce (the kinetics of) nanoparticle uptake by immune cells, to endow them with long circulation properties, and to enable them to exploit the Enhanced Permeability and Retention (EPR) effect to accumulate in tumors. More recently, with immunotherapy becoming an increasingly important cornerstone in the clinical management of cancer, ever more research efforts in academia and industry are focusing on specifically targeting immune cells with nanoparticles. In this chapter, we describe the barriers and opportunities of immune cell targeting with nanoparticles, and we discuss how nanoparticle-based drug delivery to specific immune cell populations in tumors as well as in secondary myeloid and lymphoid organs (such as bone marrow, lymph nodes, and spleen) can be leveraged to boost the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Sara Elsafy
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), Center for Biohybrid Medical Systems (CBMS), University Hospital RWTH Aachen, Aachen, Germany
| | - Josbert Metselaar
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), Center for Biohybrid Medical Systems (CBMS), University Hospital RWTH Aachen, Aachen, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), Center for Biohybrid Medical Systems (CBMS), University Hospital RWTH Aachen, Aachen, Germany.
| |
Collapse
|
23
|
Xie X, Sun Y, Peng J, Zhang Z, Wang M, Wang Z, Lei C, Huang Y, Nie Z. Collagen Anchoring Protein-Nucleic Acid Chimeric Probe for In Situ In Vivo Mapping of a Tumor-Specific Protease. Anal Chem 2023; 95:18487-18496. [PMID: 38057291 DOI: 10.1021/acs.analchem.3c03775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
In situ analysis of biomarkers in the tumor microenvironment (TME) is important to reveal their potential roles in tumor progression and early diagnosis of tumors but remains a challenge. In this work, a bottom-up modular assembly strategy was proposed for a multifunctional protein-nucleic chimeric probe (PNCP) for in situ mapping of cancer-specific proteases. PNCP, containing a collagen anchoring module and a target proteolysis-responsive isothermal amplification sensor module, can be anchored in the collagen-rich TME and respond to the target protease in situ and generate amplified signals through rolling cycle amplification of tandem fluorescent RNAs. Taking matrix metalloproteinase 2 (MMP-2), a tumor-associated protease, as the model, the feasibility of PNCP was demonstrated for the in situ detection of MMP-2 activity in 3D tumor spheroids. Moreover, in situ in vivo mapping of MMP-2 activity was also achieved in a metastatic solid tumor model with high sensitivity, providing a useful tool for evaluating tumor metastasis and distinguishing highly aggressive forms of tumors.
Collapse
Affiliation(s)
- Xuan Xie
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082, P. R. China
| | - Yuan Sun
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082, P. R. China
| | - Jialong Peng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082, P. R. China
| | - Zhenhua Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082, P. R. China
| | - Meixia Wang
- College of Biology, Hunan University, Changsha 410082, P. R. China
| | - Zeyuan Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082, P. R. China
| | - Chunyang Lei
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082, P. R. China
| | - Yan Huang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082, P. R. China
| | - Zhou Nie
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082, P. R. China
| |
Collapse
|
24
|
Kang S, Mansurov A, Kurtanich T, Chun HR, Slezak AJ, Volpatti LR, Chang K, Wang T, Alpar AT, Refvik KC, Hansen OI, Borjas GJ, Shim HN, Hultgren KT, Gomes S, Solanki A, Ishihara J, Swartz MA, Hubbell JA. Engineered IL-7 synergizes with IL-12 immunotherapy to prevent T cell exhaustion and promote memory without exacerbating toxicity. SCIENCE ADVANCES 2023; 9:eadh9879. [PMID: 38019919 PMCID: PMC10686557 DOI: 10.1126/sciadv.adh9879] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023]
Abstract
Cancer immunotherapy is moving toward combination regimens with agents of complementary mechanisms of action to achieve more frequent and robust efficacy. However, compared with single-agent therapies, combination immunotherapies are associated with increased overall toxicity because the very same mechanisms also work in concert to enhance systemic inflammation and promote off-tumor toxicity. Therefore, rational design of combination regimens that achieve improved antitumor control without exacerbated toxicity is a main objective in combination immunotherapy. Here, we show that the combination of engineered, tumor matrix-binding interleukin-7 (IL-7) and IL-12 achieves remarkable anticancer effects by activating complementary pathways without inducing any additive immunotoxicity. Mechanistically, engineered IL-12 provided effector properties to T cells, while IL-7 prevented their exhaustion and boosted memory formation as assessed by tumor rechallenge experiments. The dual combination also rendered checkpoint inhibitor (CPI)-resistant genetically engineered melanoma model responsive to CPI. Thus, our approach provides a framework of evaluation of rationally designed combinations in immuno-oncology and yields a promising therapy.
Collapse
Affiliation(s)
- Seounghun Kang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Aslan Mansurov
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Trevin Kurtanich
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Hye Rin Chun
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Anna J. Slezak
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Lisa R. Volpatti
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Kevin Chang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Thomas Wang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Aaron T. Alpar
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Kirsten C. Refvik
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - O. Isabella Hansen
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Gustavo J. Borjas
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Ha-Na Shim
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Kevin T. Hultgren
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Suzana Gomes
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Ani Solanki
- Animal Resource Center, University of Chicago, Chicago, IL, USA
| | - Jun Ishihara
- Department of Bioengineering, Imperial College London, London, UK
| | - Melody A. Swartz
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
- Committee on Cancer Biology, University of Chicago, Chicago, IL, USA
| | - Jeffrey A. Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
- Committee on Cancer Biology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
25
|
Momin N. Balancing safety and efficacy: tuning the biodistribution and pharmacokinetics of cytokine immunotherapies. Curr Opin Biotechnol 2023; 84:102994. [PMID: 37806081 DOI: 10.1016/j.copbio.2023.102994] [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: 02/26/2023] [Revised: 06/12/2023] [Accepted: 08/27/2023] [Indexed: 10/10/2023]
Abstract
Modulating the immune system shows promise in treating various conditions such as autoimmune, malignant, inflammatory, and infectious diseases. While immunotherapies can provide significant clinical benefits, they can also trigger debilitating immune-related toxicities. Achieving a balance between safety and efficacy of immunotherapy remains a significant engineering challenge. A complex immune response can be simplified into a sequence of coordinated signals with precise spatial and temporal arrangements. Mimicking or inhibiting these signals with protein immunotherapies relies on engineering them with specific biodistribution and pharmacokinetic properties. This review summarizes principles governing the movement of therapeutic proteins (i.e. biologics), focusing on cytokine immunotherapies injected intravenously or locally, and highlights approaches and considerations to balance their efficacy and safety.
Collapse
Affiliation(s)
- Noor Momin
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Center for Precision Engineering for Health, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
26
|
Silver AB, Tzeng SY, Lager M, Wang J, Ishihara J, Green JJ, Spangler JB. An engineered immunocytokine with collagen affinity improves the tumor bioavailability, tolerability, and therapeutic efficacy of IL-2. Cell Rep Med 2023; 4:101289. [PMID: 37992685 PMCID: PMC10694763 DOI: 10.1016/j.xcrm.2023.101289] [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: 06/05/2023] [Revised: 08/25/2023] [Accepted: 10/19/2023] [Indexed: 11/24/2023]
Abstract
The clinical utility of human interleukin-2 (hIL-2) is limited by its short serum half-life, preferential activation of regulatory T (TReg) over immune effector cells, and dose-limiting toxicities. We previously engineered F10 immunocytokine (IC), an intramolecularly assembled cytokine/antibody fusion protein that linked hIL-2 to an anti-IL-2 antibody (denoted F10) that extended IL-2 half-life and augmented the immune effector to TReg ratio. Here, we leveraged molecular engineering to improve the anti-tumor therapeutic efficacy and tolerability of F10 IC by developing an iteration, denoted F10 IC-CBD (collagen binding domain), designed for intratumoral administration and in situ retention based on collagen affinity. F10 IC-CBD retained IL-2 bioactivity exclusively in the tumor and eliminated IL-2-associated toxicities. Furthermore, F10 IC exhibited potent single-agent therapeutic efficacy and synergy with systemic immune checkpoint blockade and elicited an abscopal response in mouse tumors models. This engineered fusion protein presents a prototype for the design of intratumoral therapies.
Collapse
Affiliation(s)
- Aliyah B Silver
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Stephany Y Tzeng
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Mallory Lager
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical and Biomedical Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA
| | - Jeremy Wang
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Biomedical Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA
| | - Jun Ishihara
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| | - Jordan J Green
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Chemical and Biomedical Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA; Institute for NanoBioTechnology, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA; Department of Materials Science and Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jamie B Spangler
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Chemical and Biomedical Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA; Institute for NanoBioTechnology, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA; Department of Materials Science and Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
27
|
Tian F, Zhou S, Xie S, Zhang Z, Peng L, Jiang L, Wang Z, Nie Z, Huang Y. A collagen-immobilized nanodevice for in situ ratiometric imaging of cancer biomarkers in the tumor microenvironment. Chem Sci 2023; 14:12182-12193. [PMID: 37969575 PMCID: PMC10631208 DOI: 10.1039/d3sc03972b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/30/2023] [Indexed: 11/17/2023] Open
Abstract
Monitoring the spatiotemporal dynamics of cancer biomarkers within the tumor microenvironment (TME) is critical to understanding their roles in tumorigenesis. Here, we reported a multifunctional fusion protein (collagen-binding domain and duck circovirus tag fused to mCherry, CBD-mCherry-DCV) capable of binding collagen with high affinity and covalently binding specific nucleic acids with exceptional efficiency. We then constructed a chimeric protein-nucleic acid nanodevice (CPNN) using CBD-mCherry-DCV and an aptamer-based sensing module to enable spatially controlled ratiometric imaging of cancer biomarkers in the TME. The collagen-anchoring module CBD-mCherry-DCV allowed specific immobilization of CPNN on 3D multicellular tumor spheroids, enabling the sensing module to achieve "off-on" fluorescence imaging of cancer biomarkers upon specific target recognition by an aptamer. Taking advantage of the constant fluorescence signal of mCherry and the activatable fluorescence response of Cy5 to specific cancer biomarkers, the detection sensitivity and reliability of CPNN were improved by self-calibrating the signal intensity. Specifically, CPNN enabled ratiometric fluorescence imaging of varying concentrations of exogenous PDGF-BB and ATP in tumor spheroids with a high signal-to-background ratio. Furthermore, it allowed the visual monitoring of endogenous PDGF-BB and ATP released from cells. Overall, this study demonstrates the potential of the nanodevice as a versatile approach for the visualization and imaging of cancer biomarkers in the TME.
Collapse
Affiliation(s)
- Fengyu Tian
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University Changsha 410082 P. R. China
| | - Shurui Zhou
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University Changsha 410082 P. R. China
| | - Shiyi Xie
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University Changsha 410082 P. R. China
| | - Zhenhua Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University Changsha 410082 P. R. China
| | - Ling Peng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University Changsha 410082 P. R. China
| | - Ling Jiang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University Changsha 410082 P. R. China
| | - Zeyuan Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University Changsha 410082 P. R. China
| | - Zhou Nie
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University Changsha 410082 P. R. China
| | - Yan Huang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University Changsha 410082 P. R. China
| |
Collapse
|
28
|
Liu J, Xu T, Pan D, Fan J, Fu Y, Huang X, Zhao W, Dong X, Zhang S, Kuerban K, Huang X, Wang S, Chen H, He Y, Zhu YZ, Wang C, Ye L. A collagen-binding SIRPαFc fusion protein for targeted cancer immunotherapy. Int Immunopharmacol 2023; 124:110951. [PMID: 37722258 DOI: 10.1016/j.intimp.2023.110951] [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: 07/05/2023] [Revised: 09/06/2023] [Accepted: 09/13/2023] [Indexed: 09/20/2023]
Abstract
Collagen is abundant but exposed in tumor due to the abnormal tumor blood vessels, thus is considered as a tumor-specific target. The A3 domain of von Willebrand factor (vWF A3) is a kind of collagen-binding domain (CBD) which could bind collagen specifically. Previously we reported a chemosynthetic CBD-SIRPαFc conjugate, which could block CD47 and derived tumor-targeting ability by CBD. CBD-SIRPαFc conjugate represented improved anti-tumor efficacy with increased MHC II+ M1 macrophages, but the uncertain coupling ratio remained a problem. Herein, we produced a vWF A3-SIRPαFc fusion protein through eukaryotic expression system. It was examined at both molecular and cellular levels with its collagen affinity, uninfluenced original affinity to targets and phagocytosis-promoting function compared to unmodified SIRPαFc. Living imaging showed that vWF A3-SIRPαFc fusion protein derived the improved accumulation and retention in tumor than SIRPαFc. In the MC38 allograft model, vWF A3-SIRPαFc demonstrated a superior tumor-suppressing effect, characterized by increased MHC II+ M1 macrophages and T cells (particularly CD4+ T cells). These results revealed that vWF A3-SIRPαFc fusion protein derived tumor-targeting ability, leading to improved anti-tumor immunotherapeutic efficacy compared to SIRPαFc. Altogether, vWF A3 improved the anti-tumor efficacy and immune-activating function of SIRPαFc, supporting targeting tumor collagen as a possible targeted strategy.
Collapse
Affiliation(s)
- Jiayang Liu
- Minhang Hospital & Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai 201100, China
| | - Tongyang Xu
- Minhang Hospital & Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai 201100, China
| | - Danjie Pan
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China; Minhang Hospital & Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai 201100, China
| | - Jiajun Fan
- Minhang Hospital & Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai 201100, China
| | - Yuan Fu
- Minhang Hospital & Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai 201100, China
| | - Xiting Huang
- Minhang Hospital & Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai 201100, China
| | - Weili Zhao
- Minhang Hospital & Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai 201100, China
| | - Xiaochun Dong
- Minhang Hospital & Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai 201100, China
| | - Shaohui Zhang
- Minhang Hospital & Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai 201100, China
| | - Kudelaidi Kuerban
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China; Minhang Hospital & Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai 201100, China
| | - Xuan Huang
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China; Minhang Hospital & Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai 201100, China
| | - Songna Wang
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China; Minhang Hospital & Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai 201100, China
| | - Huaning Chen
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China; Minhang Hospital & Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai 201100, China
| | - Yunpeng He
- Minhang Hospital & Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai 201100, China
| | - Yi Zhun Zhu
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Congjun Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| | - Li Ye
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China; Minhang Hospital & Department of Biological Medicines at School of Pharmacy, Fudan University, Shanghai 201100, China.
| |
Collapse
|
29
|
Santollani L, Wittrup KD. Spatiotemporally programming cytokine immunotherapies through protein engineering. Immunol Rev 2023; 320:10-28. [PMID: 37409481 DOI: 10.1111/imr.13234] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
Cytokines have long been considered promising cancer immunotherapy agents due to their endogenous role in activating and proliferating lymphocytes. However, since the initial FDA approvals of Interleukin-2 (IL-2) and Interferon-ɑ (IFNɑ) for oncology over 30 years ago, cytokines have achieved little success in the clinic due to narrow therapeutic windows and dose-limiting toxicities. This is attributable to the discrepancy between the localized, regulated manner in which cytokines are deployed endogenously versus the systemic, untargeted administration used to date in most exogenous cytokine therapies. Furthermore, cytokines' ability to stimulate multiple cell types, often with paradoxical effects, may present significant challenges for their translation into effective therapies. Recently, protein engineering has emerged as a tool to address the shortcomings of first-generation cytokine therapies. In this perspective, we contextualize cytokine engineering strategies such as partial agonism, conditional activation and intratumoral retention through the lens of spatiotemporal regulation. By controlling the time, place, specificity, and duration of cytokine signaling, protein engineering can allow exogenous cytokine therapies to more closely approach their endogenous exposure profile, ultimately moving us closer to unlocking their full therapeutic potential.
Collapse
Affiliation(s)
- Luciano Santollani
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - K Dane Wittrup
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
30
|
Pabani A, Gainor JF. Facts and Hopes: Immunocytokines for Cancer Immunotherapy. Clin Cancer Res 2023; 29:3841-3849. [PMID: 37227449 DOI: 10.1158/1078-0432.ccr-22-1837] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/26/2023] [Accepted: 05/11/2023] [Indexed: 05/26/2023]
Abstract
The clinical development of cytokines as cancer therapeutics has been limited due to significant toxicities generally observed with systemic administration. This narrow therapeutic window, together with relatively modest efficacy, has made natural cytokines unattractive drug candidates. Immunocytokines represent a class of next-generation cytokines designed to overcome the challenges associated with traditional cytokines. These agents seek to improve the therapeutic index of cytokines by using antibodies as vehicles for the targeted delivery of immunomodulatory agents within the local tumor microenvironment (TME). Various molecular formats and cytokine payloads have been studied. In this review, we provide an overview of the rationale, preclinical support, and current clinical development strategies for immunocytokines.
Collapse
Affiliation(s)
- Aliyah Pabani
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Justin F Gainor
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
31
|
Wang J, Zhang Z, Chen Y. Supramolecular immunotherapy on diversiform immune cells. J Mater Chem B 2023; 11:8347-8367. [PMID: 37563947 DOI: 10.1039/d3tb00924f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Supramolecular immunotherapy employs supramolecular materials to stimulate the immune system for inhibiting tumor cell growth and metastasis, reducing the cancer recurrence rate, and improving the quality of the patient's life. Additionally, it can lessen patient suffering and the deterioration of their illness, as well as increase their survival rate. This paper will outline the fundamentals of tumor immunotherapy based on supramolecular materials as well as its current state of development and potential applications. To be more specific, we will first introduce the basic principles of supramolecular immunotherapy, including the processes, advantages and limitations of immunotherapy, the construction of supramolecular material structures, and its benefits in treatment. Second, considering the targeting of supramolecular drugs to immune cells, we comprehensively discuss the unique advantages of applying supramolecular drugs with different types of immune cells in tumor immunotherapy. The current research advances in supramolecular immunotherapy, including laboratory research and clinical applications, are also described in detail. Finally, we reveal the tremendous promise of supramolecular materials in tumor immunotherapy, as well as discuss the opportunities and challenges that may be faced in future development.
Collapse
Affiliation(s)
- Jiahui Wang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, P. R. China.
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, P. R. China
| | - Ziyi Zhang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, P. R. China.
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, P. R. China
| | - Yueyue Chen
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, P. R. China.
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, P. R. China
| |
Collapse
|
32
|
Neeser A, Ramasubramanian R, Wang C, Ma L. Engineering enhanced chimeric antigen receptor-T cell therapy for solid tumors. IMMUNO-ONCOLOGY TECHNOLOGY 2023; 19:100385. [PMID: 37483659 PMCID: PMC10362352 DOI: 10.1016/j.iotech.2023.100385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
The early clinical success and subsequent US Food and Drug Administration approval of chimeric antigen receptor (CAR)-T cell therapy for leukemia and lymphoma affirm that engineered T cells can be a powerful treatment for hematologic malignancies. Yet this success has not been replicated in solid tumors. Numerous challenges emerged from clinical experience and well-controlled preclinical animal models must be met to enable safe and efficacious CAR-T cell therapy in solid tumors. Here, we review recent advances in bioengineering strategies developed to enhance CAR-T cell therapy in solid tumors, focusing on targeted single-gene perturbation, genetic circuits design, cytokine engineering, and interactive biomaterials. These bioengineering approaches present a unique set of tools that synergize with CAR-T cells to overcome obstacles in solid tumors and achieve robust and long-lasting therapeutic efficacy.
Collapse
Affiliation(s)
- A. Neeser
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia
| | - R. Ramasubramanian
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia
| | - C. Wang
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia
| | - L. Ma
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
33
|
Liu J, Pan D, Huang X, Wang S, Chen H, Zhu YZ, Ye L. Targeting collagen in tumor extracellular matrix as a novel targeted strategy in cancer immunotherapy. Front Oncol 2023; 13:1225483. [PMID: 37692860 PMCID: PMC10484796 DOI: 10.3389/fonc.2023.1225483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/09/2023] [Indexed: 09/12/2023] Open
Abstract
Collagen, the most abundant protein in mammal, is widely expressed in tissues and organs, as well as tumor extracellular matrix. Tumor collagen mainly accumulates in tumor stroma or beneath tumor blood vessel endothelium, and is exposed due to the fragmentary structure of tumor blood vessels. Through the blood vessels with enhanced permeability and retention (EPR) effect, collagen-binding macromolecules could easily bind to tumor collagen and accumulate within tumor, supporting tumor collagen to be a potential tumor-specific target. Recently, numerous studies have verified that targeting collagen within tumor extracellular matrix (TEM) would enhance the accumulation and retention of immunotherapy drugs at tumor, significantly improving their anti-tumor efficacy, as well as avoiding severe adverse effects. In this review, we would summarize the known collagen-binding domains (CBD) or proteins (CBP), their mechanism and application in tumor-targeting immunotherapy, and look forward to future development.
Collapse
Affiliation(s)
- Jiayang Liu
- Department of Biological Medicines at School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai, China
| | - Danjie Pan
- Department of Biological Medicines at School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai, China
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
| | - Xuan Huang
- Department of Biological Medicines at School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai, China
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
| | - Songna Wang
- Department of Biological Medicines at School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai, China
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
| | - Huaning Chen
- Department of Biological Medicines at School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai, China
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
| | - Yi Zhun Zhu
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
| | - Li Ye
- Department of Biological Medicines at School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, China
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
| |
Collapse
|
34
|
Flies DB, Langermann S, Jensen C, Karsdal MA, Willumsen N. Regulation of tumor immunity and immunotherapy by the tumor collagen extracellular matrix. Front Immunol 2023; 14:1199513. [PMID: 37662958 PMCID: PMC10470046 DOI: 10.3389/fimmu.2023.1199513] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/28/2023] [Indexed: 09/05/2023] Open
Abstract
It has been known for decades that the tumor extracellular matrix (ECM) is dysfunctional leading to loss of tissue architecture and promotion of tumor growth. The altered ECM and tumor fibrogenesis leads to tissue stiffness that act as a physical barrier to immune cell infiltration into the tumor microenvironment (TME). It is becoming increasingly clear that the ECM plays important roles in tumor immune responses. A growing body of data now indicates that ECM components also play a more active role in immune regulation when dysregulated ECM components act as ligands to interact with receptors on immune cells to inhibit immune cell subpopulations in the TME. In addition, immunotherapies such as checkpoint inhibitors that are approved to treat cancer are often hindered by ECM changes. In this review we highlight the ways by which ECM alterations affect and regulate immunity in cancer. More specifically, how collagens and major ECM components, suppress immunity in the complex TME. Finally, we will review how our increased understanding of immune and immunotherapy regulation by the ECM is leading towards novel disruptive strategies to overcome immune suppression.
Collapse
|
35
|
Ding JY, Sun L, Zhu ZH, Wu XC, Xu XL, Xiang YW. Nano drug delivery systems: a promising approach to scar prevention and treatment. J Nanobiotechnology 2023; 21:268. [PMID: 37568194 PMCID: PMC10416511 DOI: 10.1186/s12951-023-02037-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Scar formation is a common physiological process that occurs after injury, but in some cases, pathological scars can develop, leading to serious physiological and psychological effects. Unfortunately, there are currently no effective means to intervene in scar formation, and the structural features of scars and their unclear mechanisms make prevention and treatment even more challenging. However, the emergence of nanotechnology in drug delivery systems offers a promising avenue for the prevention and treatment of scars. Nanomaterials possess unique properties that make them well suited for addressing issues related to transdermal drug delivery, drug solubility, and controlled release. Herein, we summarize the recent progress made in the use of nanotechnology for the prevention and treatment of scars. We examine the mechanisms involved and the advantages offered by various types of nanomaterials. We also highlight the outstanding challenges and questions that need to be addressed to maximize the potential of nanotechnology in scar intervention. Overall, with further development, nanotechnology could significantly improve the prevention and treatment of pathological scars, providing a brighter outlook for those affected by this condition.
Collapse
Affiliation(s)
- Jia-Ying Ding
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lu Sun
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhi-Heng Zhu
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xi-Chen Wu
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiao-Ling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, 310015, PR China.
| | - Yan-Wei Xiang
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
36
|
Closset L, Gultekin O, Salehi S, Sarhan D, Lehti K, Gonzalez-Molina J. The extracellular matrix - immune microenvironment crosstalk in cancer therapy: Challenges and opportunities. Matrix Biol 2023; 121:217-228. [PMID: 37524251 DOI: 10.1016/j.matbio.2023.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/02/2023]
Abstract
Targeting the tumour immune microenvironment (TIME) by cancer immunotherapy has led to improved patient outcomes. However, response to these treatments is heterogeneous and cancer-type dependant. The therapeutic activity of classical cancer therapies such as chemotherapy, radiotherapy, and surgical oncology is modulated by alterations of the TIME. A major regulator of immune cell function and resistance to both immune and classical therapies is the extracellular matrix (ECM). Concurrently, cancer therapies reshape the TIME as well as the ECM, causing both pro- and anti-tumour responses. Accordingly, the TIME-ECM crosstalk presents attractive opportunities to improve therapy outcomes. Here, we review the molecular crosstalk between the TIME and the ECM in cancer and its implications in cancer progression and clinical intervention. Additionally, we discuss examples and future directions of ECM and TIME co-targeting in combination with oncological therapies including surgery, chemotherapy, and radiotherapy.
Collapse
Affiliation(s)
- Lara Closset
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, Stockholm 171 65, Sweden; Saint-Antoine Research center (CRSA), UMR_S 938, INSERM, Sorbonne Université, Paris F-75012, France
| | - Okan Gultekin
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, Stockholm 171 65, Sweden
| | - Sahar Salehi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, Stockholm 171 65, Sweden; Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet, Stockholm, Sweden; Department of Pelvic Cancer, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Dhifaf Sarhan
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Kaisa Lehti
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, Stockholm 171 65, Sweden; Department of Biomedical Laboratory Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jordi Gonzalez-Molina
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, Stockholm 171 65, Sweden.
| |
Collapse
|
37
|
Rana I, Oh J, Baig J, Moon JH, Son S, Nam J. Nanocarriers for cancer nano-immunotherapy. Drug Deliv Transl Res 2023; 13:1936-1954. [PMID: 36190661 PMCID: PMC9528883 DOI: 10.1007/s13346-022-01241-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2022] [Indexed: 11/03/2022]
Abstract
The host immune system possesses an intrinsic ability to target and kill cancer cells in a specific and adaptable manner that can be further enhanced by cancer immunotherapy, which aims to train the immune system to boost the antitumor immune response. Several different categories of cancer immunotherapy have emerged as new standard cancer therapies in the clinic, including cancer vaccines, immune checkpoint inhibitors, adoptive T cell therapy, and oncolytic virus therapy. Despite the remarkable survival benefit for a subset of patients, the low response rate and immunotoxicity remain the major challenges for current cancer immunotherapy. Over the last few decades, nanomedicine has been intensively investigated with great enthusiasm, leading to marked advancements in nanoparticle platforms and nanoengineering technology. Advances in nanomedicine and immunotherapy have also led to the emergence of a nascent research field of nano-immunotherapy, which aims to realize the full therapeutic potential of immunotherapy with the aid of nanomedicine. In particular, nanocarriers present an exciting opportunity in immuno-oncology to boost the activity, increase specificity, decrease toxicity, and sustain the antitumor efficacy of immunological agents by potentiating immunostimulatory activity and favorably modulating pharmacological properties. This review discusses the potential of nanocarriers for cancer immunotherapy and introduces preclinical studies designed to improve clinical cancer immunotherapy modalities using nanocarrier-based engineering approaches. It also discusses the potential of nanocarriers to address the challenges currently faced by immuno-oncology as well as the challenges for their translation to clinical applications.
Collapse
Affiliation(s)
- Isra Rana
- College of Pharmacy, Chonnam National University, Gwangju, 61186, South Korea
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Jaeeun Oh
- Department of Biological Sciences, Inha University, Incheon, 22212, South Korea
| | - Juwon Baig
- Department of Biological Sciences, Inha University, Incheon, 22212, South Korea
| | - Jeong Hyun Moon
- Department of Biological Sciences, Inha University, Incheon, 22212, South Korea
| | - Sejin Son
- Department of Biological Sciences, Inha University, Incheon, 22212, South Korea.
- Department of Biological Sciences and Bioengineering, Inha University/Industry-Academia Interactive R&E Center for Bioprocess Innovation, Inha University, Incheon, South Korea.
| | - Jutaek Nam
- College of Pharmacy, Chonnam National University, Gwangju, 61186, South Korea.
| |
Collapse
|
38
|
Bruni S, Mercogliano MF, Mauro FL, Cordo Russo RI, Schillaci R. Cancer immune exclusion: breaking the barricade for a successful immunotherapy. Front Oncol 2023; 13:1135456. [PMID: 37284199 PMCID: PMC10239871 DOI: 10.3389/fonc.2023.1135456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 05/10/2023] [Indexed: 06/08/2023] Open
Abstract
Immunotherapy has changed the course of cancer treatment. The initial steps were made through tumor-specific antibodies that guided the setup of an antitumor immune response. A new and successful generation of antibodies are designed to target immune checkpoint molecules aimed to reinvigorate the antitumor immune response. The cellular counterpart is the adoptive cell therapy, where specific immune cells are expanded or engineered to target cancer cells. In all cases, the key for achieving positive clinical resolutions rests upon the access of immune cells to the tumor. In this review, we focus on how the tumor microenvironment architecture, including stromal cells, immunosuppressive cells and extracellular matrix, protects tumor cells from an immune attack leading to immunotherapy resistance, and on the available strategies to tackle immune evasion.
Collapse
|
39
|
Wang F, Huang Q, Su H, Sun M, Wang Z, Chen Z, Zheng M, Chakroun R, Monroe M, Chen D, Wang Z, Gorelick N, Serra R, Wang H, Guan Y, Suk J, Tyler B, Brem H, Hanes J, Cui H. Self-assembling paclitaxel-mediated stimulation of tumor-associated macrophages for postoperative treatment of glioblastoma. Proc Natl Acad Sci U S A 2023; 120:e2204621120. [PMID: 37098055 PMCID: PMC10161130 DOI: 10.1073/pnas.2204621120] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 03/09/2023] [Indexed: 04/26/2023] Open
Abstract
The unique cancer-associated immunosuppression in brain, combined with a paucity of infiltrating T cells, contributes to the low response rate and poor treatment outcomes of T cell-based immunotherapy for patients diagnosed with glioblastoma multiforme (GBM). Here, we report on a self-assembling paclitaxel (PTX) filament (PF) hydrogel that stimulates macrophage-mediated immune response for local treatment of recurrent glioblastoma. Our results suggest that aqueous PF solutions containing aCD47 can be directly deposited into the tumor resection cavity, enabling seamless hydrogel filling of the cavity and long-term release of both therapeutics. The PTX PFs elicit an immune-stimulating tumor microenvironment (TME) and thus sensitizes tumor to the aCD47-mediated blockade of the antiphagocytic "don't eat me" signal, which subsequently promotes tumor cell phagocytosis by macrophages and also triggers an antitumor T cell response. As adjuvant therapy after surgery, this aCD47/PF supramolecular hydrogel effectively suppresses primary brain tumor recurrence and prolongs overall survivals with minimal off-target side effects.
Collapse
Affiliation(s)
- Feihu Wang
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Whiting School of Engineering, Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD21218
- Center for Nanomedicine, Wilmer Eye Institute, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
| | - Qian Huang
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, The Johns Hopkins University, Baltimore, MD21205
| | - Hao Su
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Whiting School of Engineering, Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD21218
| | - Mingjiao Sun
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Whiting School of Engineering, Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD21218
- Center for Nanomedicine, Wilmer Eye Institute, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
| | - Zeyu Wang
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
| | - Ziqi Chen
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Whiting School of Engineering, Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD21218
| | - Mengzhen Zheng
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Whiting School of Engineering, Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD21218
| | - Rami W. Chakroun
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Whiting School of Engineering, Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD21218
| | - Maya K. Monroe
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Whiting School of Engineering, Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD21218
| | - Daiqing Chen
- Center for Nanomedicine, Wilmer Eye Institute, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
| | - Zongyuan Wang
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Whiting School of Engineering, Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD21218
| | - Noah Gorelick
- Department of Neurosurgery, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
| | - Riccardo Serra
- Department of Neurosurgery, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
| | - Han Wang
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Whiting School of Engineering, Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD21218
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, The Johns Hopkins University, Baltimore, MD21205
- Department of Neurological Surgery, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
| | - Jung Soo Suk
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Center for Nanomedicine, Wilmer Eye Institute, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
- Department of Neurological Surgery, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
| | - Betty Tyler
- Department of Neurosurgery, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
| | - Henry Brem
- Department of Neurosurgery, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
- Department of Ophthalmology, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
- Department of Biomedical Engineering, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
| | - Justin Hanes
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Center for Nanomedicine, Wilmer Eye Institute, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
- Department of Ophthalmology, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
- Department of Biomedical Engineering, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Whiting School of Engineering, Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD21218
- Center for Nanomedicine, Wilmer Eye Institute, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
- Department of Materials Science and Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
| |
Collapse
|
40
|
Manna S, Maiti S, Shen J, Weiss A, Mulder E, Du W, Esser-Kahn AP. Nanovaccine that activates the NLRP3 inflammasome enhances tumor specific activation of anti-cancer immunity. Biomaterials 2023; 296:122062. [PMID: 36863071 PMCID: PMC10085859 DOI: 10.1016/j.biomaterials.2023.122062] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023]
Abstract
Neoantigen cancer vaccines that target tumor specific mutations are emerging as a promising modality for cancer immunotherapy. To date, various approaches have been adopted to enhance efficacy of these therapies, but the low immunogenicity of neoantigens has hindered clinical application. To address this challenge, we developed a polymeric nanovaccine platform that activates the NLRP3 inflammasome, a key immunological signaling pathway in pathogen recognition and clearance. The nanovaccine is comprised of a poly (orthoester) scaffold engrafted with a small-molecule TLR7/8 agonist and an endosomal escape peptide that facilitates lysosomal rupture and NLRP3 inflammasome activation. Upon solvent transfer, the polymer self-assembles with neoantigens to form ∼50 nm nanoparticles that facilitate co-delivery to antigen-presenting cells. This polymeric activator of the inflammasome (PAI) was found to induce potent antigen-specific CD8+ T cell responses characterized by IFN-γ and GranzymeB secretion. Moreover, in combination with immune checkpoint blockade therapy, the nanovaccine stimulated robust anti-tumor immune responses against established tumors in EG.7-OVA, B16·F10, and CT-26 models. Results from our studies indicate that NLRP3 inflammasome activating nanovaccines demonstrate promise for development as a robust platform to enhance immunogenicity of neoantigen therapies.
Collapse
Affiliation(s)
- Saikat Manna
- Pritzker School of Molecular Engineering, University of Chicago, 5640 S. Ellis Ave., Chicago, IL 60637, USA
| | - Sampa Maiti
- Pritzker School of Molecular Engineering, University of Chicago, 5640 S. Ellis Ave., Chicago, IL 60637, USA; Department of Chemistry and Biochemistry, Science of Advanced Material, Central Michigan University, Mount Pleasant, MI 48858, United States
| | - Jingjing Shen
- Pritzker School of Molecular Engineering, University of Chicago, 5640 S. Ellis Ave., Chicago, IL 60637, USA
| | - Adam Weiss
- Pritzker School of Molecular Engineering, University of Chicago, 5640 S. Ellis Ave., Chicago, IL 60637, USA; Department of Chemistry, University of Chicago, 5735 S Ellis Ave., Chicago, IL 60637, USA
| | - Elizabeth Mulder
- Pritzker School of Molecular Engineering, University of Chicago, 5640 S. Ellis Ave., Chicago, IL 60637, USA
| | - Wenjun Du
- Department of Chemistry and Biochemistry, Science of Advanced Material, Central Michigan University, Mount Pleasant, MI 48858, United States
| | - Aaron P Esser-Kahn
- Pritzker School of Molecular Engineering, University of Chicago, 5640 S. Ellis Ave., Chicago, IL 60637, USA.
| |
Collapse
|
41
|
Bohmer M, Xue Y, Jankovic K, Dong Y. Advances in engineering and delivery strategies for cytokine immunotherapy. Expert Opin Drug Deliv 2023; 20:579-595. [PMID: 37104673 PMCID: PMC10330431 DOI: 10.1080/17425247.2023.2208344] [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: 11/01/2022] [Accepted: 04/25/2023] [Indexed: 04/29/2023]
Abstract
INTRODUCTION Cytokine immunotherapy is a growing field for the treatment of cancer, infectious disease, autoimmunity, and other ailments. Therapeutic cytokines are a class of secreted, small proteins that play a pivotal role in regulating the innate and adaptive immune system by provoking or mitigating immune responses. In the clinic, cytokines are frequently combined with other treatments, such as small molecules and monoclonal antibodies. However, the clinical translation of cytokine therapies is hindered by their short half-life, pleiotropic nature, and off-target effects, which cause diminished efficacy and severe systemic toxicity. Such toxicity limits dosage, thus resulting in suboptimal doses. Accordingly, numerous efforts have been devoted to exploring strategies to promote cytokine therapies by improving their tissue specificity and pharmacokinetics. AREAS COVERED Preclinical and clinical research into bioengineering and delivery strategies for cytokines, consisting of bioconjugation, fusion proteins, nanoparticles, and scaffold-based systems. EXPERT OPINION These approaches pave the way for the development of next-generation cytokine treatments with greater clinical benefit and reduced toxicity, circumventing such issues currently associated with cytokine therapy.
Collapse
Affiliation(s)
- Margaret Bohmer
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Yonger Xue
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Katarina Jankovic
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
- Pelotonia Institute for Immune-Oncology, The Ohio State University, Columbus, OH, 43210, USA
- Center for Cancer Engineering, The Ohio State University, Columbus, OH, 43210, USA
- Center for Cancer Metabolism, Department of Radiation Oncology, The Ohio State University, Columbus, OH, 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
- The Center for Clinical and Translational Science, The Ohio State University, Columbus, OH, 43210, USA
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
- Icahn Genomics Institute, Precision Immunology Institute, Department of Oncological Sciences, Tisch Cancer Institute, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| |
Collapse
|
42
|
Bhatia V, Kamat NV, Pariva TE, Wu LT, Tsao A, Sasaki K, Sun H, Javier G, Nutt S, Coleman I, Hitchcock L, Zhang A, Rudoy D, Gulati R, Patel RA, Roudier MP, True LD, Srivastava S, Morrissey CM, Haffner MC, Nelson PS, Priceman SJ, Ishihara J, Lee JK. Targeting advanced prostate cancer with STEAP1 chimeric antigen receptor T cell and tumor-localized IL-12 immunotherapy. Nat Commun 2023; 14:2041. [PMID: 37041154 PMCID: PMC10090190 DOI: 10.1038/s41467-023-37874-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 04/04/2023] [Indexed: 04/13/2023] Open
Abstract
Six transmembrane epithelial antigen of the prostate 1 (STEAP1) is a cell surface antigen for therapeutic targeting in prostate cancer. Here, we report broad expression of STEAP1 relative to prostate-specific membrane antigen (PSMA) in lethal metastatic prostate cancers and the development of a STEAP1-directed chimeric antigen receptor (CAR) T cell therapy. STEAP1 CAR T cells demonstrate reactivity in low antigen density, antitumor activity across metastatic prostate cancer models, and safety in a human STEAP1 knock-in mouse model. STEAP1 antigen escape is a recurrent mechanism of treatment resistance and is associated with diminished tumor antigen processing and presentation. The application of tumor-localized interleukin-12 (IL-12) therapy in the form of a collagen binding domain (CBD)-IL-12 fusion protein combined with STEAP1 CAR T cell therapy enhances antitumor efficacy by remodeling the immunologically cold tumor microenvironment of prostate cancer and combating STEAP1 antigen escape through the engagement of host immunity and epitope spreading.
Collapse
Affiliation(s)
- Vipul Bhatia
- Human Biology Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Nikhil V Kamat
- Division of Medical Oncology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Tiffany E Pariva
- Human Biology Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Li-Ting Wu
- Human Biology Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Annabelle Tsao
- Human Biology Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Koichi Sasaki
- Department of Bioengineering, Imperial College London, 86 Wood Lane, London, W12 0BZ, UK
| | - Huiyun Sun
- Human Biology Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Gerardo Javier
- Human Biology Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Sam Nutt
- Human Biology Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Ilsa Coleman
- Human Biology Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Lauren Hitchcock
- Human Biology Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Ailin Zhang
- Human Biology Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Dmytro Rudoy
- Human Biology Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Roman Gulati
- Public Health Sciences Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Radhika A Patel
- Human Biology Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Martine P Roudier
- Department of Urology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Lawrence D True
- Department of Pathology and Laboratory Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Shivani Srivastava
- Human Biology Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Colm M Morrissey
- Department of Urology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Michael C Haffner
- Human Biology Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
- Department of Pathology and Laboratory Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Peter S Nelson
- Human Biology Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
- Division of Medical Oncology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
- Department of Urology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
- Department of Pathology and Laboratory Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Saul J Priceman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, USA
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Jun Ishihara
- Department of Bioengineering, Imperial College London, 86 Wood Lane, London, W12 0BZ, UK.
| | - John K Lee
- Human Biology Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA.
- Division of Medical Oncology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA.
- Department of Pathology and Laboratory Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA.
- Clinical Research Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA.
| |
Collapse
|
43
|
Wang Y, Li W, Li Z, Mo F, Chen Y, Iida M, Wheeler DL, Hu Q. Active recruitment of anti-PD-1-conjugated platelets through tumor-selective thrombosis for enhanced anticancer immunotherapy. SCIENCE ADVANCES 2023; 9:eadf6854. [PMID: 36989364 PMCID: PMC10058243 DOI: 10.1126/sciadv.adf6854] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 03/01/2023] [Indexed: 06/19/2023]
Abstract
Immune checkpoint inhibitors (ICIs) can reinvigorate T cells to eradicate tumor cells, showing great potential in combating various types of tumors. We propose a delivery strategy to enhance tumor-selective ICI accumulation, which leverages the responsiveness of platelets and platelet-derivatives to coagulation cascade signals. A fused protein tTF-RGD targets tumor angiogenic blood vessel endothelial cells and initiates the coagulation locoregionally at the tumor site, forming a "cellular hive" to recruit anti-PD-1 antibody (aPD-1)-conjugated platelets to the tumor site and subsequently activating platelets to release aPD-1 antibody to reactivate T cells for improved immunotherapy. Moreover, on a patient-derived xenograft breast cancer model, the platelet membrane-coated nanoparticles can also respond to the coagulation signals initiated by tTF-RGD, thus enhancing the accumulation and antitumor efficacy of the loaded chemotherapeutics. Our study illustrates a versatile platform technology to enhance the local accumulation of ICIs and chemodrugs by taking advantage of the responsiveness of platelets and platelet derivatives to thrombosis.
Collapse
Affiliation(s)
- Yixin Wang
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Wen Li
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Zhaoting Li
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Fanyi Mo
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yu Chen
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Mari Iida
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA
| | - Deric L Wheeler
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
44
|
Palmeri JR, Lax BM, Peters JM, Duhamel L, Stinson JA, Santollani L, Lutz EA, Pinney W, Bryson BD, Wittrup KD. Tregs constrain CD8 + T cell priming required for curative intratumorally anchored anti-4-1BB immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.30.526116. [PMID: 36778460 PMCID: PMC9915483 DOI: 10.1101/2023.01.30.526116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Although co-stimulation of T cells with agonist antibodies targeting 4-1BB (CD137) improves antitumor immune responses in preclinical studies, clinical development has been hampered by on-target, off-tumor toxicity. Here, we report the development of a tumor-anchored ɑ4-1BB agonist (ɑ4-1BB-LAIR), which consists of an ɑ4-1BB antibody fused to the collagen binding protein LAIR. While combination treatment with an antitumor antibody (TA99) displayed only modest efficacy, simultaneous depletion of CD4+ T cells boosted cure rates to over 90% of mice. We elucidated two mechanisms of action for this synergy: ɑCD4 eliminated tumor draining lymph node Tregs, enhancing priming and activation of CD8+ T cells, and TA99 + ɑ4-1BB-LAIR supported the cytotoxic program of these newly primed CD8+ T cells within the tumor microenvironment. Replacement of ɑCD4 with ɑCTLA-4, a clinically approved antibody that enhances T cell priming, produced equivalent cure rates while additionally generating robust immunological memory against secondary tumor rechallenge.
Collapse
Affiliation(s)
- Joseph R Palmeri
- Koch Institute for Integrative Cancer Research; Cambridge, MA
- Department of Chemical Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
| | - Brianna M Lax
- Koch Institute for Integrative Cancer Research; Cambridge, MA
- Department of Chemical Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
| | - Joshua M Peters
- Department of Biological Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
- Ragon Institute of MIT, MGH, and Harvard; Cambridge, MA
| | - Lauren Duhamel
- Koch Institute for Integrative Cancer Research; Cambridge, MA
- Department of Biological Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
| | - Jordan A Stinson
- Koch Institute for Integrative Cancer Research; Cambridge, MA
- Department of Biological Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
| | - Luciano Santollani
- Koch Institute for Integrative Cancer Research; Cambridge, MA
- Department of Chemical Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
| | - Emi A Lutz
- Koch Institute for Integrative Cancer Research; Cambridge, MA
- Department of Biological Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
| | - William Pinney
- Koch Institute for Integrative Cancer Research; Cambridge, MA
- Department of Biological Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
| | - Bryan D Bryson
- Department of Biological Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
- Ragon Institute of MIT, MGH, and Harvard; Cambridge, MA
| | - K Dane Wittrup
- Koch Institute for Integrative Cancer Research; Cambridge, MA
- Department of Chemical Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
- Department of Biological Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
| |
Collapse
|
45
|
Deckers J, Anbergen T, Hokke AM, de Dreu A, Schrijver DP, de Bruin K, Toner YC, Beldman TJ, Spangler JB, de Greef TFA, Grisoni F, van der Meel R, Joosten LAB, Merkx M, Netea MG, Mulder WJM. Engineering cytokine therapeutics. NATURE REVIEWS BIOENGINEERING 2023; 1:286-303. [PMID: 37064653 PMCID: PMC9933837 DOI: 10.1038/s44222-023-00030-y] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Cytokines have pivotal roles in immunity, making them attractive as therapeutics for a variety of immune-related disorders. However, the widespread clinical use of cytokines has been limited by their short blood half-lives and severe side effects caused by low specificity and unfavourable biodistribution. Innovations in bioengineering have aided in advancing our knowledge of cytokine biology and yielded new technologies for cytokine engineering. In this Review, we discuss how the development of bioanalytical methods, such as sequencing and high-resolution imaging combined with genetic techniques, have facilitated a better understanding of cytokine biology. We then present an overview of therapeutics arising from cytokine re-engineering, targeting and delivery, mRNA therapeutics and cell therapy. We also highlight the application of these strategies to adjust the immunological imbalance in different immune-mediated disorders, including cancer, infection and autoimmune diseases. Finally, we look ahead to the hurdles that must be overcome before cytokine therapeutics can live up to their full potential.
Collapse
Affiliation(s)
- Jeroen Deckers
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Medical Centre, Nijmegen, Netherlands
| | - Tom Anbergen
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Medical Centre, Nijmegen, Netherlands
| | - Ayla M. Hokke
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Anne de Dreu
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - David P. Schrijver
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Koen de Bruin
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Yohana C. Toner
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Medical Centre, Nijmegen, Netherlands
| | - Thijs J. Beldman
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Medical Centre, Nijmegen, Netherlands
| | - Jamie B. Spangler
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD USA
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Tom F. A. de Greef
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands
- Centre for Living Technologies, Alliance Eindhoven University of Technology, Wageningen University & Research, Utrecht University and University Medical Center Utrecht (EWUU), Utrecht, Netherlands
| | - Francesca Grisoni
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
- Centre for Living Technologies, Alliance Eindhoven University of Technology, Wageningen University & Research, Utrecht University and University Medical Center Utrecht (EWUU), Utrecht, Netherlands
| | - Roy van der Meel
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Present Address: Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Leo A. B. Joosten
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Medical Centre, Nijmegen, Netherlands
- Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, Nijmegen, Netherlands
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Maarten Merkx
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Present Address: Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Medical Centre, Nijmegen, Netherlands
- Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, Nijmegen, Netherlands
- Department for Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Willem J. M. Mulder
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Medical Centre, Nijmegen, Netherlands
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Present Address: Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
46
|
George A, Varghese J, Padinharayil H. Potential of Biotechnology in Cancer Management. NOVEL TECHNOLOGIES IN BIOSYSTEMS, BIOMEDICAL & DRUG DELIVERY 2023:9-44. [DOI: 10.1007/978-981-99-5281-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
47
|
Prospect of bacteria for tumor diagnosis and treatment. Life Sci 2022; 312:121215. [PMID: 36414093 DOI: 10.1016/j.lfs.2022.121215] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/21/2022]
Abstract
In recent decades, the comprehensive cancer treatments including surgery, chemotherapy, and radiotherapy have improved the overall survival rate and quality of life of many cancer patients. However, we are still facing many difficult problems in the cancer treatment, such as unpredictable side effects, high recurrence rate, and poor curative effect. Therefore, the better intervention strategies are needed in this field. In recent years, the role and importance of microbiota in a variety of diseases were focused on as a hot research topic, and the role of some intracellular bacteria of cancer cells in carcinogenesis has recently been discovered. The impact of bacteria on cancer is not limited to their contribution to tumorigenesis, but the overall susceptibility of bacteria to subsequent tumor progression, the development of concurrent infections, and the response to anti-cancer therapy have also been found to be affected. Concerns about the contribution of bacteria in the anti-cancer response have inspired researchers to develop bacteria-based anti-cancer treatments. In this paper, we reviewed the main roles of bacteria in the occurrence and development of tumors, and summarized the mechanism of bacteria in the occurrence, development, and clinical anti-tumor treatment of tumors, providing new insights for the in-depth study of the role of bacteria in tumor diagnosis and treatment. This review aims to provide a new perspective for the development of new technologies based on bacteria to enhance anti-tumor immunotherapy.
Collapse
|
48
|
Kirchhammer N, Trefny MP, Auf der Maur P, Läubli H, Zippelius A. Combination cancer immunotherapies: Emerging treatment strategies adapted to the tumor microenvironment. Sci Transl Med 2022; 14:eabo3605. [DOI: 10.1126/scitranslmed.abo3605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Immune checkpoint blockade (ICB) has revolutionized cancer treatment. However, resistance to ICB occurs frequently due to tumor-intrinsic alterations or extrinsic factors in the tumor microenvironment. This Viewpoint aims to give an update on recent developments in immunotherapy for solid tumors and highlights progress in translational research and clinical practice.
Collapse
Affiliation(s)
- Nicole Kirchhammer
- Cancer Immunology, Department of Biomedicine, University and University Hospital Basel, Basel 4031, Switzerland
| | - Marcel P. Trefny
- Cancer Immunology, Department of Biomedicine, University and University Hospital Basel, Basel 4031, Switzerland
| | - Priska Auf der Maur
- Tumor Heterogeneity, Metastasis and Resistance, Department of Biomedicine, University and University Hospital of Basel, Basel 4031, Switzerland
| | - Heinz Läubli
- Cancer Immunotherapy, Department of Biomedicine, University and University Hospital Basel, Basel 4031, Switzerland
- Medical Oncology, University Hospital Basel, Basel 4031, Switzerland
| | - Alfred Zippelius
- Cancer Immunology, Department of Biomedicine, University and University Hospital Basel, Basel 4031, Switzerland
- Medical Oncology, University Hospital Basel, Basel 4031, Switzerland
| |
Collapse
|
49
|
Peng H, He X, Wang Q. Targeted drug delivery system for ovarian cancer microenvironment: Improving the effects of immunotherapy. Front Immunol 2022; 13:1035997. [PMID: 36405688 PMCID: PMC9670735 DOI: 10.3389/fimmu.2022.1035997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
Immunotherapies have shown modest benefits in the current clinical trials for ovarian cancer. The tumor microenvironment (TME) in an immunosuppressive phenotype contributes to this “failure” of immunotherapy in ovarian cancer. Many stromal cell types in the TME (e.g., tumor-associated macrophages and fibroblasts) have been identified as having plasticity in pro- and antitumor activities and are responsible for suppressing the antitumor immune response. Thus, the TME is an extremely valuable target for adjuvant interventions to improve the effects of immunotherapy. The current strategies targeting the TME include: 1) eliminating immunosuppressive cells or transforming them into immunostimulatory phenotypes and 2) inhibiting their immunosuppressive or pro-tumor production. Most of the effective agents used in the above strategies are genetic materials (e.g., cDNA, mRNA, or miRNA), proteins, or other small molecules (e.g., peptides), which are limited in their target and instability. Various formulations of drug delivery system (DDS) have been designed to realize the controlled release and targeting delivery of these agents to the tumor sites. Nanoparticles and liposomes are the most frequently exploited materials. Based on current evidence from preclinical and clinical studies, the future of the DDS is promising in cancer immunotherapy since the combination of agents with a DDS has shown increased efficacy and decreased toxicities compared with free agents. In the future, more efforts are needed to further identify the hallmarks and biomarkers in the ovarian TME, which is crucial for the development of more effective, safe, and personalized DDSs.
Collapse
|
50
|
Baldari S, Di Modugno F, Nisticò P, Toietta G. Strategies for Efficient Targeting of Tumor Collagen for Cancer Therapy. Cancers (Basel) 2022; 14:cancers14194706. [PMID: 36230627 PMCID: PMC9563908 DOI: 10.3390/cancers14194706] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/25/2022] Open
Abstract
Simple Summary The tumor microenvironment encompasses the cellular and extracellular matrix components that support and shape the three-dimensional framework in which solid tumors develop and grow. The extracellular matrix of the tumor is characterized by increased deposition and aberrant architecture of collagen fibers. Therefore, as a key mechanical component of the tumor microenvironment, collagen plays a critical role in cancer progression, metastasis, and therapeutic response. To boost the efficacy of current anticancer therapies, including immunotherapy, innovative approaches should take into account strategies directed against the dysregulated non-cancer cell stromal components. In the current review, we provide an overview of the principal approaches to target tumor collagen to provide therapeutic benefits. Abstract The tumor stroma, which comprises stromal cells and non-cellular elements, is a critical component of the tumor microenvironment (TME). The dynamic interactions between the tumor cells and the stroma may promote tumor progression and metastasis and dictate resistance to established cancer therapies. Therefore, novel antitumor approaches should combine anticancer and anti-stroma strategies targeting dysregulated tumor extracellular matrix (ECM). ECM remodeling is a hallmark of solid tumors, leading to extensive biochemical and biomechanical changes, affecting cell signaling and tumor tissue three-dimensional architecture. Increased deposition of fibrillar collagen is the most distinctive alteration of the tumor ECM. Consequently, several anticancer therapeutic strategies have been developed to reduce excessive tumor collagen deposition. Herein, we provide an overview of the current advances and challenges of the main approaches aiming at tumor collagen normalization, which include targeted anticancer drug delivery, promotion of degradation, modulation of structure and biosynthesis of collagen, and targeting cancer-associated fibroblasts, which are the major extracellular matrix producers.
Collapse
|