• HOME
  • NEWS
  • EXPLORE
    • CAREER
      • Companies
      • Jobs
    • EVENTS
    • iGEM
      • News
      • Team
    • PHOTOS
    • VIDEO
    • WIKI
  • BLOG
  • COMMUNITY
    • FACEBOOK
    • INSTAGRAM
    • TWITTER
Saturday, July 26, 2025
BIOENGINEER.ORG
No Result
View All Result
  • Login
  • HOME
  • NEWS
  • EXPLORE
    • CAREER
      • Companies
      • Jobs
        • Lecturer
        • PhD Studentship
        • Postdoc
        • Research Assistant
    • EVENTS
    • iGEM
      • News
      • Team
    • PHOTOS
    • VIDEO
    • WIKI
  • BLOG
  • COMMUNITY
    • FACEBOOK
    • INSTAGRAM
    • TWITTER
  • HOME
  • NEWS
  • EXPLORE
    • CAREER
      • Companies
      • Jobs
        • Lecturer
        • PhD Studentship
        • Postdoc
        • Research Assistant
    • EVENTS
    • iGEM
      • News
      • Team
    • PHOTOS
    • VIDEO
    • WIKI
  • BLOG
  • COMMUNITY
    • FACEBOOK
    • INSTAGRAM
    • TWITTER
No Result
View All Result
Bioengineer.org
No Result
View All Result
Home NEWS Science News Cancer

Calibr-Skaggs Administers First Patient with Switchable CAR-T Cell Therapy in Phase 1 Trial Targeting Metastatic Breast Cancer

Bioengineer by Bioengineer
June 17, 2025
in Cancer
Reading Time: 5 mins read
0
ADVERTISEMENT
Share on FacebookShare on TwitterShare on LinkedinShare on RedditShare on Telegram

blank

Revolutionizing Solid Tumor Treatment: The Dawn of Switchable CAR-T Therapy in Advanced Breast Cancer

In a groundbreaking advancement poised to reshape cancer therapy, researchers at the Calibr-Skaggs Institute for Innovative Medicines, part of the renowned Scripps Research, have initiated a first-in-human clinical trial evaluating a novel switchable chimeric antigen receptor T cell (sCAR-T) therapy for advanced breast cancer. This Phase 1 dose-escalation study—identified as NCT06878248—explores the safety, tolerability, pharmacokinetics, and pharmacodynamics of a combination therapy comprising CLBR001, an engineered autologous T cell product, and ABBV-461, an antibody-based biologic acting as a molecular “switch.” This initiative marks the pioneering application of the sCAR-T platform in the treatment of solid tumors, a frontier long plagued by therapeutic challenges.

Traditional CAR-T therapies have profoundly altered the landscape of hematological malignancy treatment, delivering curative potential for patients with refractory blood cancers. Despite these successes, their translation to solid tumors like breast cancer has been hindered by the complex and immunosuppressive tumor microenvironment, antigen heterogeneity, and safety concerns related to on-target off-tumor effects. The Calibr-Skaggs sCAR-T platform innovatively addresses these obstacles by embedding modularity and control into the CAR-T design, potentially empowering clinicians with precision on par with a remote control system.

.adsslot_wHQ5oalZ7W{ width:728px !important; height:90px !important; }
@media (max-width:1199px) { .adsslot_wHQ5oalZ7W{ width:468px !important; height:60px !important; } }
@media (max-width:767px) { .adsslot_wHQ5oalZ7W{ width:320px !important; height:50px !important; } }

ADVERTISEMENT

The sCAR-T approach integrates an engineered T cell component, CLBR001, with an antibody-based “switch” molecule, ABBV-461. This switch bridges the T cells and tumor antigens, selectively activating the cytotoxic T cells only in the presence of the switch antibody. Through this mechanism, dosing of the switch can be externally modulated, offering unprecedented temporal control over CAR-T activity, which may minimize the risk of adverse events such as cytokine release syndrome, neurotoxicity, or immune exhaustion. Consequently, it paves the way for safer and more effective CAR-T interventions in the notoriously resilient solid tumor milieu.

Preclinical and early clinical evidence has underscored the therapeutic potential of this design. Unlike conventional CAR-T cells, CLBR001 cells have demonstrated the capacity for robust in vivo expansion within hostile tumor microenvironments, a critical factor in overcoming solid tumor resistance. Moreover, the ability to intermittently “switch off” these engineered cells supports their functional longevity by mitigating exhaustion, a state of diminished T cell efficacy associated with chronic antigen exposure. These features collectively suggest that sCAR-T therapy may surmount key biological barriers that have previously limited CAR-T effectiveness outside hematological settings.

Travis Young, PhD, vice president of biology at Calibr-Skaggs, highlights the significance of this innovation: “There’s a critical need to develop gene and cell therapy approaches that are able to recreate the success observed in blood cancers for patients with solid tumors like breast cancer. By integrating an antibody-based ‘switch,’ there’s the potential to enhance the precision of targeting solid tumor cells, while also mitigating potential safety risks.” This perspective emphasizes the dual focus on efficacy and safety, a balance essential for moving cell-based therapies into mainstream solid tumor oncology.

The ongoing Phase 1 trial is structured as an open-label, dose-escalation study enrolling patients with locally advanced or metastatic breast cancer who have exhausted standard treatment options and lack alternatives. Participants receive a single infusion of CLBR001 cells subsequent to lymphodepletion, a regimen that conditions the immune system for the engraftment and expansion of infused T cells. Subsequently, patients undergo successive cycles of ABBV-461 administration, with meticulous monitoring to delineate the optimal dosing parameters, safety profile, and preliminary efficacy signals.

Mechanistically, the switch molecule ABBV-461 binds simultaneously to the tumor antigen and the engineered receptor on CLBR001 cells. This bifunctional interaction acts akin to a molecular toggle, enabling selective activation of CAR-T cells in the tumor vicinity, thereby sparing healthy tissues. Such controllability introduces a versatile therapeutic window, allowing clinicians to fine-tune treatment intensity in real time or transiently cease switch administration in response to adverse events. This sophisticated control mechanism addresses one of the long-standing challenges in CAR-T therapy: managing unpredictable toxicities without compromising antitumor potency.

This trial also represents a notable collaboration between Calibr-Skaggs and AbbVie, combining cutting-edge cell therapy engineering with advanced biologics expertise. Their partnership exemplifies the translational synergy necessary to shepherd pioneering immunotherapies from bench to bedside, accelerating the availability of innovative treatments that target unmet medical needs.

Beyond this specific clinical endeavor, Calibr-Skaggs delineates its sCAR-T platform as a transformative paradigm within the broader scope of immuno-oncology. The platform’s modular design permits adaptability across various tumor antigens and cancer types, potentially enabling customizable regimens tailored to individual patient tumor profiles. Such flexibility is crucial in heterogeneous cancers like breast carcinoma, where intra- and inter-patient variability often confound standardized treatments.

Scripps Research, the parent institution, continues to stand at the forefront of biomedical innovation, with an ecosystem that nurtures fundamental discovery, translational medicine, and interdisciplinary collaboration. Ranked among the world’s most influential research entities, Scripps fosters integration across genomics, digital health, and informatics—all instrumental in refining patient stratification and enhancing therapeutic outcomes. The Calibr-Skaggs initiative exemplifies this multi-dimensional approach, leveraging sophisticated cellular engineering within a patient-centric framework.

The introduction of switchable CAR-T therapy to the challenging realm of solid tumors heralds a new chapter in cancer immunotherapy. By marrying precise molecular control with the potent cytotoxic machinery of T cells, this strategy aspires to transform refractory breast cancer from a formidable adversary into a manageable condition with durable responses. While early-stage trials must confirm safety and define optimal dosing, the scientific rationale and preliminary data offer compelling optimism for patients who currently face limited options.

In the rapidly evolving landscape of immuno-oncology, the sCAR-T platform’s emphasis on controllability and adaptability may set a benchmark for future cell therapies. It underscores an emerging ethos where therapeutic efficacy is harmonized with safety through sophisticated bioengineering, enhancing not only treatment outcomes but also patient quality of life. As this trial unfolds, it will be closely watched by the scientific community and clinicians alike for insights that may unlock the full potential of T cell therapies against solid tumors.

With this clinical trial underway, the oncology field eagerly anticipates data that could redefine therapeutic paradigms for breast cancer and beyond. Should CLBR001 + ABBV-461 demonstrate acceptable safety and encouraging efficacy, the sCAR-T technology could catalyze a wave of modular, controllable cell therapies addressing various hard-to-treat solid malignancies, pushing the boundaries of personalized medicine and heralding a new era in cancer therapeutics.

Subject of Research: Development and clinical evaluation of switchable CAR-T cell therapy (sCAR-T) for advanced and metastatic breast cancer.

Article Title: Revolutionizing Solid Tumor Treatment: The Dawn of Switchable CAR-T Therapy in Advanced Breast Cancer

News Publication Date: (Not specified in the source)

Web References:

Clinical Trial NCT06878248: http://clinicaltrials.gov/study/NCT06878248
Calibr-Skaggs Institute: https://calibr.scripps.edu/
Scripps Research: http://www.scripps.edu

Keywords: Breast cancer, solid tumors, CAR-T therapy, switchable CAR-T, sCAR-T, immunotherapy, CLBR001, ABBV-461, cellular therapy, cancer treatment, T cell exhaustion, tumor microenvironment

Tags: ABBV-461 antibody biologicadvanced solid tumor therapyCalibr-Skaggs Institute researchcancer immunotherapy innovationschimeric antigen receptor technologyengineered autologous T cell therapymetastatic breast cancer treatmentPhase 1 clinical trialprecision medicine in oncologysafety and tolerability in cancer trialsswitchable CAR-T cell therapytherapeutic challenges in solid tumors

Share12Tweet8Share2ShareShareShare2

Related Posts

blank

Hepatoblastoma Trends: Dynamic SDI Analysis

July 5, 2025
Noninvasive Nasopharyngeal Cancer Detection via Gene Methylation

Noninvasive Nasopharyngeal Cancer Detection via Gene Methylation

July 5, 2025

Molecular Biomarkers Predicting Adult Glioma Radiosensitivity

July 5, 2025

Aerobic Exercises Combat Fatigue in Colorectal Cancer

July 5, 2025

POPULAR NEWS

  • Blind to the Burn

    Overlooked Dangers: Debunking Common Myths About Skin Cancer Risk in the U.S.

    62 shares
    Share 25 Tweet 16
  • AI Achieves Breakthrough in Drug Discovery by Tackling the True Complexity of Aging

    70 shares
    Share 28 Tweet 18
  • USF Research Unveils AI Technology for Detecting Early PTSD Indicators in Youth Through Facial Analysis

    43 shares
    Share 17 Tweet 11
  • Dr. Miriam Merad Honored with French Knighthood for Groundbreaking Contributions to Science and Medicine

    46 shares
    Share 18 Tweet 12

About

We bring you the latest biotechnology news from best research centers and universities around the world. Check our website.

Follow us

Recent News

Additive Manufacturing of Monolithic Gyroidal Solid Oxide Cells

Machine Learning Uncovers Sorghum’s Complex Mold Resistance

Pathology Multiplexing Revolutionizes Disease Mapping

  • Contact Us

Bioengineer.org © Copyright 2023 All Rights Reserved.

Welcome Back!

Login to your account below

Forgotten Password?

Retrieve your password

Please enter your username or email address to reset your password.

Log In
No Result
View All Result
  • Homepages
    • Home Page 1
    • Home Page 2
  • News
  • National
  • Business
  • Health
  • Lifestyle
  • Science

Bioengineer.org © Copyright 2023 All Rights Reserved.