Dr Hadas Weinstein-Marom

Dr Hadas Weinstein-Marom
Principal Researcher
Research Group Leader
PhD
Research Interests:

Head of the Laboratory for Cellular Immunotherapy & Synthetic Biology at MIGAL – Galilee Research Institute in Kiryat Shmona, Israel.

Gene-based cellular immunotherapy has been my main field of interest throughout my entire scientific career. I carried out my Ph.D. research at the Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel, under the joint supervision of Profs. Tamar Peretz, Michal Lotem and Gideon Gross. My study focused on the development of new membrane-attached derivatives of several key cytokines in the context of adoptive T cell therapy of cancer. I conducted postdoctoral research in Tel Aviv University at the Ella Lemelbaum Institute for Immuno-Oncology, Chaim Sheba Medical Center, Israel. My research focused on the development of a new gene-based approach for adoptive T cell therapy of cancer, which combines several strategies in this field.

In the field of immuno-gene therapy of cancer I focus on improving selectivity and safety of adoptive T cell therapy and on enhancing the functional properties and survival of antitumor T cells. In the field of inflammatory diseases I concentrate on genetically engineering regulatory T cells as a new tool for the treatment of inflammatory diseases. Following is a brief description of the main directions of my scientific work.

  • Membrane-Attached Cytokines for T Cell Therapy

During my Ph.D., I developed a strategy to enhance the survival and function of adoptive T cells by expressing key cytokines (IL-2, IL-12, and IL-15) as integral T-cell membrane proteins. Systemic administration of these cytokines is often associated with severe toxicity, limiting their clinical use. By attaching them to the cell membrane via mRNA electroporation, I confined cytokine availability to therapeutic T cells, preventing unwanted systemic effects.

The engineered cytokines supported the ex vivo growth of human CD8+ and CD4+ T cells for six days post-transfection, demonstrating comparable effects to high-dose soluble IL-2. Membrane-bound cytokines also enhanced activation markers such as CD25, CD69, 4-1BB, and OX40 when combined with TLR4 signaling. These findings support the potential application of membrane cytokines in improving adoptive T cell therapy.

  • Synergy Between Membrane Cytokines and Genetic Adjuvants in T Cell Therapy

Building on my Ph.D. research, I investigated the cooperative effects of membrane cytokines and other genetic adjuvants, such as constitutively active TLR4 and CD40. These genetic modifications were designed to autonomously enhance T cell function upon mRNA electroporation.

In both peripheral blood CD8+ T cells and tumor-infiltrating lymphocytes (TILs), these engineered adjuvants significantly upregulated key activation markers (CD25, OX40, 4-1BB, CD127, CD28), improved cytotoxic activity, and increased IFN-γ and TNF-α secretion. Notably, predefined mRNA mixtures encoding these adjuvants synergistically enhanced melanoma-specific T cell responses, demonstrating their potential in cancer immunotherapy.

  • Optimizing Retroviral Transduction of Tumor-Infiltrating Lymphocytes (TILs)

During my postdoctoral research, I developed an optimized protocol for retroviral transduction of TILs, allowing for efficient genetic modification to improve their therapeutic efficacy. Given the resurgence of TIL therapy due to its ability to target tumor-specific neoantigens, this advancement was critical in ensuring high transduction efficiency and functional enhancement of T cells.

Using this protocol, we achieved >70% expression of an anti-CD19 chimeric antigen receptor (CAR) in TILs, resulting in strong IFN-γ secretion in response to CD19+ target cells. This protocol provides a reproducible approach to enhancing TIL function for clinical applications.

  • CD40 as a Costimulatory Element in CAR-T Cell Therapy

As a principal investigator, I led a study exploring the use of CD40 as a novel costimulatory domain in second- and third-generation CAR-T cells. The costimulatory domain in CAR constructs plays a crucial role in regulating T cell activation, persistence, and anti-tumor efficacy.

We compared CD40 to the widely used 4-1BB domain in CAR constructs. Our results showed that CD40 triggered potent NF-κB activation and was comparable to 4-1BB in enhancing T cell activation markers, cytokine secretion, and cytotoxicity. Notably, donor-dependent differences in response to CD40 and 4-1BB suggested a potential for personalized approaches in CAR-T cell design. These findings highlight CD40 as a promising alternative costimulatory domain in next-generation CAR therapies.

In two ongoing translational studies carried out with our clinician partners in Israel we are assessing new second and third generation CARs harboring the CD40 element for the treatment of B cell malignancies and melanoma.

  • Membrane-Anchored IL-18-Based Genetic Adjuvants for Cancer Immunotherapy

I initiated and led the development of a composite genetic adjuvant that integrates IL-18, TLR4, and CD40 into a single membrane-bound construct. The goal was to amplify T cell activation by simultaneously engaging the cytokine receptor, toll-like receptor, and TNF receptor pathways.

This "all-in-one" genetic modification was tested in CAR-T cells and melanoma patient-derived TILs. Engineered T cells exhibited spontaneous upregulation of T-bet, increased IFN-γ and TNF-α secretion, and enhanced melanoma cell killing. Notably, the IL-18-TLR4-CD40 construct was more potent than membrane IL-18 alone, demonstrating the value of integrating multiple costimulatory signals into a single construct. This approach provides a novel way to improve adoptive T cell therapy outcomes while avoiding the systemic toxicity associated with cytokine administration.

  • Implementation of new genetic switches and logic gates in adoptive T cell therapy of cancer

We are currently exploring various gene circuits we have been developing in recent years in order to address different challenges in cancer immunotherapy:

· Lack of tumor-specific antigens

· On-target off-tumor toxicity of CAR-T cell therapy

· Tumor escape from TCR recognition following loss of antigen presentation

· Tumor escape from CAR recognition via antigen loss

In these studies we are examining new OR, AND and NOT modules, specifically designed to tackle each of these challenges in the context of several human cancers.

  • Generation of Human Type 1 Regulatory T (Tr1)-Like Cells via Membrane IL-10

As part of my work as a PI, I developed a method for generating Tr1-like cells through membrane-attached IL-10 rather than soluble IL-10. Tr1 cells are FoxP3-negative regulatory T cells that secrete IL-10 and play a role in immune suppression. Their therapeutic use requires efficient protocols for stable and reproducible generation.

Using retroviral transduction, I successfully engineered human CD4+ T cells to express membrane-bound IL-10, leading to stable IL-10 expression and allowing for simple purification using magnetic beads. Purified cells exhibited a homogenous Tr1-like phenotype (CD49b+, LAG-3+, PD-1+, pSTAT3+) and maintained a central memory phenotype. This approach overcomes the limitations of uncontrolled IL-10 secretion and offers a promising strategy for generating regulatory T cells for immune modulation.

Scientific Publications

No publications found