Liv-1 Targeting Antibody Drug Conjugate

Introduction

In this article, we describe a novel antibody-drug conjugate (ADC; SGN-LIV1A), targeting the zinc transporter LIV-1 (SLC39A6) for the treatment of metastatic breast cancer.​ LIV-1 was previously known to be expressed by estrogen receptor-positive breast cancers.​.​.​

Description of Liv-1 Targeting Antibody Drug Conjugate

In this article, a novel antibody-drug conjugate (ADC; SGN-LIV1A) targeting the zinc transporter LIV-1 (SLC39A6) for metastatic breast cancer treatment is discussed.​ LIV-1, a transmembrane protein expressed in various cancer types, is the focus of this ADC. The investigational ADC, SGN-LIV1A, comprises a humanized IgG1 antibody linked to monomethyl auristatin E (MMAE).​ This ADC is designed to target LIV-1 on cell surfaces, inducing potent cytotoxic cell killing.​ The promising potential of SGN-LIV1A as a therapeutic agent for LIV-1-expressing cancers is highlighted…​

Background Information

Liv-1, a zinc transporter expressed in various cancers, is targeted by the investigational ADC SGN-LIV1A.​ This ADC comprises a humanized IgG1 antibody linked to MMAE for potent cell killing.​

Liv-1 Protein and Its Role in Breast Cancer

Liv-1, a zinc transporter, is highly prevalent in breast cancer cells.​ The investigational ADC SGN-LIV1A targets Liv-1 using a humanized IgG1 monoclonal antibody conjugated to monomethyl auristatin E (MMAE).​ Liv-1 is expressed in various cancer types and plays a critical role in metastatic progression.​ The expression of Liv-1 is maintained after hormonal therapy, making it a promising target for ADC therapy in breast cancer patients.​

Ladiratuzumab Vedotin (LV) as an Investigational ADC

Ladiratuzumab vedotin (LV), also known as SGN-LIV1A, is an investigational antibody-drug conjugate (ADC) developed to target the zinc transporter LIV-1 (SLC39A6) in various cancer types.​ This novel ADC combines a humanized IgG1 monoclonal antibody with monomethyl auristatin E (MMAE) via a protease-cleavable linker.​ When LV binds to LIV-1 on cell surfaces, it triggers internalization and the release of MMAE, leading to potent cytotoxic cell killing.​ LV shows promising potential as a targeted therapy for cancers expressing LIV-1.

Clinical Trials and Development

The Phase I clinical trial of BRY812, a novel antibody-drug conjugate (ADC) that targets LIV-1, has been initiated for advanced malignant tumors.​ This third-generation ADC shows promise as a targeted therapy for treating various cancer types.​

Phase I Clinical Trial of BRY812 for Advanced Malignant Tumors

BioRay Pharmaceutical Co.​, Ltd. has initiated a Phase I clinical trial for BRY812, a third-generation antibody-drug conjugate (ADC) targeting LIV-1 for advanced malignant tumors.​ The trial, conducted at Sun Yat-sen Memorial Hospital, marks a significant advancement in targeted cancer therapy.​

Acceptance of BRY812 by China NMPA for Clinical Trial

BRY812, a novel antibody-drug conjugate (ADC) targeting LIV-1, has been accepted by the China National Medical Products Administration (NMPA) for a clinical trial.​ This acceptance marks a significant step in the development of targeted therapies against advanced malignant tumors.​

Advancements and Future Prospects

The development of Liv-1 targeting antibody-drug conjugates (ADCs) like SGN-LIV1A presents exciting prospects for the treatment of metastatic breast cancer.​ Engineering technologies continue to enhance ADC optimization٫ offering novel targets and innovative strategies to improve efficacy in solid tumors.​

Role of Liv-1 as a Promising Target for ADCs

Liv-1, a zinc transporter, serves as a promising target for antibody-drug conjugates (ADCs) in cancer therapy.​ The expression of Liv-1 in various cancer types, including triple-negative breast cancer, highlights its potential as a therapeutic target.​ ADCs like SGN-LIV1A, specifically designed to target Liv-1, offer a targeted approach for cancer treatment with reduced systemic toxicity and potent antitumor effects.​

Engineering Technologies and Novel Targets for ADC Optimization

Constant advancements in engineering technologies are enhancing the optimization of antibody-drug conjugates (ADCs) targeting Liv-1.​ These technologies aim to improve the efficacy of ADCs by introducing novel targets and innovative strategies for optimal drug delivery and cytotoxic effects in solid tumors.​ The evolution of engineering techniques is crucial for refining the precision and therapeutic potential of ADCs in cancer treatment.​

10 responses to “Liv-1 Targeting Antibody Drug Conjugate”

  1. Nora Avatar
    Nora

    The potential of SGN-LIV1A in targeting LIV-1 opens up new possibilities for improving outcomes in breast cancer patients.

  2. Hannah Avatar
    Hannah

    The role of zinc transporter targeting in breast cancer therapy is a unique and innovative approach.

  3. Sophia Avatar
    Sophia

    Exciting advancements in breast cancer treatment! Looking forward to seeing the impact of this novel antibody-drug conjugate.

  4. Aiden Avatar
    Aiden

    The concept of using antibody-drug conjugates like SGN-LIV1A is a fascinating development in cancer therapy.

  5. Isabella Avatar
    Isabella

    Intriguing approach to utilize zinc transporter targeting for treating metastatic breast cancer.

  6. Max Avatar
    Max

    The development of SGN-LIV1A as an ADC for metastatic breast cancer showcases the progress in precision medicine.

  7. Leo Avatar
    Leo

    The description of SGN-LIV1A as an antibody-drug conjugate for breast cancer treatment is a significant advancement.

  8. Lily Avatar
    Lily

    The specificity of SGN-LIV1A for LIV-1 in breast cancer treatment is a step towards personalized medicine.

  9. Ethan Avatar
    Ethan

    The targeting of LIV-1 with SGN-LIV1A seems promising for estrogen receptor-positive breast cancers.

  10. Owen Avatar
    Owen

    This article sheds light on the potential of targeted therapy for metastatic breast cancer with the ADC SGN-LIV1A.

Leave a Reply

Your email address will not be published. Required fields are marked *