Cellular Immunotherapies for Acute Lymphoblastic Leukemia (ALL) represent a cutting-edge advancement in hematologic oncology and regenerative immunotherapy, redefining the treatment landscape for this aggressive hematological malignancy. ALL, a cancer of the lymphoid line of blood cells, is characterized by the overproduction of immature lymphoblasts, leading to bone marrow failure and systemic complications. Despite the success of chemotherapy protocols, radiation, and stem cell transplantation, challenges remain—particularly for relapsed and refractory cases. DrStemCellsThailand (DRSCT) now harnesses advanced cellular immunotherapy platforms to target leukemic cells with unprecedented precision, offering hope beyond traditional therapies [1-3].
Conventional ALL treatment approaches, though often effective in pediatric populations, face significant limitations in adult and high-risk patients. Chemotherapeutic toxicity, drug resistance, and immunosuppression can impair quality of life and long-term remission. Moreover, these treatments largely aim to eliminate cancerous cells without reconstituting the patient’s immune surveillance or hematopoietic balance. These shortcomings underscore the critical need for strategies that restore immune function, eliminate minimal residual disease (MRD), and reprogram the immune system for long-term surveillance.
At the frontier of regenerative medicine, Cellular Immunotherapies for Acute Lymphoblastic Leukemia (ALL)—including CAR-T cell therapy, NK cell infusions, and TCR-engineered lymphocytes—represent a paradigm shift in the treatment of ALL. These strategies utilize genetically modified autologous or allogeneic immune cells to recognize and destroy leukemic blasts while sparing healthy tissue. Imagine a future where a patient’s own immune system is re-engineered to hunt down leukemia cells, maintain remission, and prevent relapse. This vision is becoming a reality at DRSCT, where innovation meets immune precision. Join us as we explore this revolutionary frontier, where immunoengineering converges with regenerative science to redefine what is possible in the treatment of ALL [1-3].
At the Anti-Aging and Regenerative Medicine Center of Thailand, our hematology and genomics team offers comprehensive genetic testing to assess predisposition and optimize the efficacy of Cellular Immunotherapies for ALL. While ALL often arises from somatic mutations, inherited susceptibilities and treatment responsiveness can vary significantly between individuals. Understanding each patient’s genetic blueprint is pivotal in designing effective immunotherapeutic strategies.
Our genomic profiling includes analysis of mutations in IKZF1, TP53, PAX5, CDKN2A/B, and chromosomal translocations such as BCR-ABL1, ETV6-RUNX1, and MLL rearrangements—all of which affect leukemogenesis, prognosis, and immunotherapy response. Moreover, pharmacogenomic insights into thiopurine methyltransferase (TPMT) and NUDT15 polymorphisms allow us to personalize adjunct chemotherapy when needed [1-3].
We also investigate human leukocyte antigen (HLA) profiles and immunogenomic landscapes to optimize donor matching and reduce graft-versus-host disease (GVHD) risk in allogeneic settings. Understanding tumor antigen heterogeneity, such as expression of CD19, CD22, and FLT3, enables precise selection of immunotherapeutic targets, such as CAR-T constructs or bispecific T-cell engagers (BiTEs).
This genomic-guided, precision medicine approach enhances patient selection, predicts adverse events, and maximizes therapeutic outcomes, ultimately serving as the foundation for tailored Cellular Immunotherapies for Acute Lymphoblastic Leukemia (ALL) regimens. Armed with this knowledge, our team crafts individualized treatment blueprints to optimize success rates while minimizing risks [1-3].
Acute Lymphoblastic Leukemia (ALL) arises from a malignant transformation of lymphoid progenitor cells, primarily affecting the bone marrow and lymphatic system. Its pathogenesis is a multistep process involving genetic mutations, epigenetic dysregulation, disrupted signaling pathways, and impaired immune surveillance. Below is an in-depth look at the pathobiology that underpins ALL and the therapeutic opportunities for cellular immunotherapy:
Leukemogenic Mutations and Chromosomal Rearrangements
Clonal Expansion and Escape from Apoptosis
Niche Hijacking
Immune Suppression and Checkpoint Dysregulation
Minimal Residual Disease (MRD) and Relapse
Central Nervous System Infiltration
Resistance Mechanisms
Chimeric Antigen Receptor T-Cell Therapy (CAR-T)
Natural Killer (NK) Cell Therapies
T-Cell Receptor (TCR) Engineering
BiTE Antibodies and Immune Checkpoint Inhibitors
Conclusion
The pathogenesis of ALL involves a sophisticated network of genetic lesions, immune evasion tactics, and microenvironmental interactions that collectively promote leukemic survival and proliferation. Cellular Immunotherapies for Acute Lymphoblastic Leukemia (ALL) offer a transformative approach—one that not only targets these malignant mechanisms but also restores immune competence, reprograms the hematopoietic niche, and offers durable remission. By targeting the disease at a cellular and molecular level, we move beyond palliation to potential cures [1-3].
Acute Lymphoblastic Leukemia (ALL) is a rapidly progressing hematologic malignancy characterized by uncontrolled proliferation of immature lymphoid precursors in the bone marrow, blood, and extramedullary tissues. The etiology of ALL is multifactorial, involving genetic mutations, epigenetic dysregulation, immunologic failure, and environmental insults. Key pathophysiologic mechanisms include:
Genetic Mutations and Chromosomal Rearrangements
The leukemogenic transformation of lymphoid progenitor cells is often driven by chromosomal abnormalities such as:
These mutations disrupt normal hematopoiesis and drive uncontrolled proliferation of leukemic blasts.
Bone Marrow Microenvironment Dysregulation
The leukemic niche undergoes profound changes, including:
This altered microenvironment facilitates immune escape and contributes to therapeutic resistance [4-7].
Epigenetic Dysregulation and Transcriptional Rewiring
Aberrant DNA methylation, histone modifications, and non-coding RNA expression contribute to leukemogenesis by:
These epigenetic alterations often co-occur with genetic lesions, reinforcing leukemic cell survival and proliferation.
Immunologic Dysfunctions and Immune Evasion
ALL blasts can manipulate immune signaling by:
These strategies contribute to immune escape and disease persistence.
Environmental and Iatrogenic Factors
Although genetic factors are primary, environmental contributors include:
Understanding the interplay between these mechanisms is critical to designing targeted, cell-based immunotherapeutic strategies for ALL [4-7].
Despite improved survival in pediatric ALL, conventional treatment of ALL—especially in adults—remains fraught with limitations. Current chemotherapeutic regimens rely on cytotoxic agents and stem cell transplantation but face numerous barriers:
Chemoresistance and Relapse
Toxicity and Quality of Life
Limitations of Hematopoietic Stem Cell Transplantation (HSCT)
Inability to Harness Long-Term Immune Surveillance
These challenges underscore the need for durable, targeted, and immune-mediated interventions such as Cellular Immunotherapies for Acute Lymphoblastic Leukemia (ALL) [4-7].
Cellular Immunotherapies for Acute Lymphoblastic Leukemia (ALL) has revolutionized the treatment landscape for ALL, particularly in relapsed/refractory (R/R) settings. The most impactful breakthroughs involve engineered immune cells designed to target leukemic blasts with high specificity and persistence:
CAR T Cell Therapy (Chimeric Antigen Receptor T Cells)
Year: 2017
Researcher: Dr. Stephan Grupp
Institution: Children’s Hospital of Philadelphia (CHOP), USA
Result: FDA approval of Tisagenlecleucel (Kymriah) for pediatric and young adult R/R B-ALL.
CAR T cells engineered to target CD19 achieved 90% complete remission in initial trials. Patients demonstrated long-term remission with reconstituted immune surveillance.
Dual-Targeted CAR T Cells (CD19/CD22)
Year: 2020
Researcher: Dr. Renier Brentjens
Institution: Memorial Sloan Kettering Cancer Center, USA
Result: Dual antigen targeting reduced relapse rates caused by antigen escape. Showed 73% durable remission in high-risk R/R B-ALL patients.
Allogeneic (“Off-the-Shelf”) CAR T Cell Therapy
Year: 2021
Researcher: Dr. Marcela Maus
Institution: Massachusetts General Hospital, USA
Result: Universal CAR T cells from healthy donors using TALEN or CRISPR editing demonstrated safety and efficacy in early-phase ALL trials, overcoming logistical and cost barriers [4-7].
CAR-NK Cell Therapy (Natural Killer Cells)
Year: 2022
Researcher: Dr. Katy Rezvani
Institution: MD Anderson Cancer Center, USA
Result: CD19-CAR NK cells from umbilical cord blood induced remission in 70% of patients without cytokine release syndrome (CRS) or GVHD, offering a safer alternative to T cells.
γδ T Cell Therapy
Year: 2023
Researcher: Dr. Michael Hudecek
Institution: University of Würzburg, Germany
Result: Preclinical models showed γδ T cells with engineered TCRs targeting intracellular leukemia-associated antigens eliminated leukemic blasts across diverse HLA backgrounds.
TCR-Engineered T Cell Therapy
Year: 2024
Researcher: Dr. Stanley Riddell
Institution: Fred Hutchinson Cancer Center, USA
Result: T cells with high-affinity TCRs targeting Wilms tumor antigen-1 (WT1) successfully reduced MRD and prolonged survival in post-HSCT ALL settings.
Extracellular Vesicle (EV)-Mediated Immunomodulation
Year: 2025
Researcher: Dr. Cecilia Lindefors
Institution: Karolinska Institute, Sweden
Result: CAR-T derived EVs containing CD19-targeting nanobodies and cytokine cargo induced targeted apoptosis in leukemic blasts while re-educating immune suppressive cells.
These landmark innovations are redefining ALL treatment paradigms, offering precision targeting, immune memory, and reduced systemic toxicity, and creating new hope for high-risk patients [4-7].
Several public figures and medical advocates have helped raise awareness for ALL and promoted the exploration of regenerative and immune-based therapies:
These figures play a critical role in humanizing the science and accelerating funding and research momentum for next-generation cellular therapies in ALL [4-7].
Acute Lymphoblastic Leukemia (ALL) is driven by complex cellular dysregulation in the bone marrow and lymphoid tissues, characterized by unchecked proliferation of lymphoid progenitors. Understanding the key cellular components affected in ALL opens the door for targeted cellular immunotherapies:
Leukemic Blasts: Malignant lymphoid progenitor cells (usually B-lineage, less frequently T-lineage) that crowd out normal hematopoiesis and cause bone marrow failure. These blasts evade apoptosis and exhibit clonal expansion due to mutations in transcription factors and signaling pathways (e.g., NOTCH1, TEL-AML1, BCR-ABL).
T Cells: Central to immune surveillance. In ALL, cytotoxic T cells become dysfunctional or exhausted, impairing their ability to clear leukemic cells. Restoring their function is a major goal of immunotherapy.
Regulatory T Cells (Tregs): Typically suppress immune overactivation, but in ALL, Tregs can be overrepresented, protecting leukemic cells from immune attack by suppressing cytotoxic responses.
Natural Killer (NK) Cells: Innate immune effectors with potent anti-leukemic potential. In ALL, NK cell activity is often reduced or functionally impaired, limiting natural immune clearance.
Mesenchymal Stromal Cells (MSCs): Present in the bone marrow niche, MSCs interact with leukemic cells and can unintentionally support leukemic survival by secreting anti-apoptotic factors and remodeling the stromal microenvironment.
Dendritic Cells (DCs): Key antigen-presenting cells that orchestrate T cell responses. Their function is often blunted in ALL, reducing effective T-cell priming and limiting immune clearance of leukemic cells.
CAR-T Cells: Genetically engineered T cells directed against surface markers like CD19 on leukemic blasts. They represent a revolutionary advance, enabling selective cytotoxicity with memory response potential.
By targeting these dysregulated cellular interactions, Cellular Immunotherapies for Acute Lymphoblastic Leukemia (ALL) seeks not just remission but long-term immune-mediated control or eradication of the disease [8-10].
Progenitor Stem Cells (PSC) of Lymphoid Lineage
PSC of T Cells: For generation of naive and cytotoxic T cells that can be redirected or reprogrammed against leukemic targets.
PSC of NK Cells: For expansion of high-potency NK cells with anti-leukemia cytotoxic profiles.
PSC of Dendritic Cells: Enabling the reconstitution of functional antigen-presenting machinery in immunosuppressed ALL patients.
PSC of Mesenchymal Stromal Cells: Modulating the marrow microenvironment, potentially rewiring stromal signals to suppress leukemic niches.
PSC of Anti-Leukemia Immune Cells: Such as γδ T cells and invariant NKT cells, with innate anti-leukemic capacities.
PSC of Hematopoietic Stem Cells (HSCs): Foundational for reconstitution of healthy hematopoiesis post-myeloablative therapy [8-10].
Our specialized immunotherapeutic strategies utilize Progenitor Stem Cells (PSCs) to address cellular deficits and immune escape mechanisms characteristic of ALL:
Lymphoid Progenitors: Healthy PSC-derived lymphoid progenitors help re-establish normal hematopoiesis and suppress leukemic dominance.
T Cell Progenitors: Engineered or naïve T cells are programmed to develop into functional cytotoxic T lymphocytes targeting ALL-specific antigens (e.g., CD19, CD22).
NK Cell Progenitors: Expanded and matured ex vivo to yield NK cells with superior tumor lytic capacity and antibody-dependent cytotoxicity.
Dendritic Cell Progenitors: Used to restore or enhance antigen presentation to break ALL-induced immune tolerance.
Stromal Reprogramming Cells: PSC-derived MSCs engineered to secrete leukemia-suppressive cytokines (e.g., IL-15, IFN-γ) while preserving marrow support.
Fibrosis and Inflammation-Modulating Cells: Addressing bone marrow fibrosis and inflammatory damage induced by leukemic infiltration or chemotherapy.
Harnessing this cellular diversity through progenitor-based immunotherapy allows for a comprehensive, multi-targeted assault on ALL at its molecular and cellular roots [8-10].
At DrStemCellsThailand (DRSCT)’s Immuno-Oncology Center, we employ ethically sourced allogeneic cells with high immunologic and regenerative potential:
Umbilical Cord Blood-Derived HSCs: Enable full hematopoietic reconstitution with reduced graft-versus-host disease (GVHD) risk in post-chemotherapy settings.
Cord Blood-Derived NK Cells: Readily expanded and activated for adoptive NK cell therapy in relapsed/refractory ALL.
Wharton’s Jelly MSCs: Provide immunosuppressive control during transplantation and modulate Treg populations for GVHD prevention.
Placental-Derived Immune Cells: Rich in naïve T cells and NK progenitors, offering unique anti-leukemic activity.
Bone Marrow-Derived T and NK Cell Precursors: Used for ex vivo expansion and genetic modification into tumor-specific effector cells (e.g., CAR-T cells).
These allogeneic platforms enable scalable, off-the-shelf options for personalized and population-wide Cellular Immunotherapy for ALL [8-10].
Initial Recognition of ALL Pathophysiology: Dr. Thomas Hodgkin, 1832
One of the earliest recognitions of lymphoid malignancy pathology, laying foundational work for modern leukemia classification.
Discovery of BCR-ABL Fusion in Leukemia: Dr. Janet Rowley, 1973
Chromosomal translocations in leukemia were first identified, linking cytogenetics to targeted therapies for lymphoid leukemias, including Ph+ ALL.
First Bone Marrow Transplant in Leukemia: Dr. E. Donnall Thomas, 1957–1977
Nobel-winning development of bone marrow transplantation, now integral in ALL consolidation therapy.
CAR-T Cell Breakthrough: Dr. Carl June, University of Pennsylvania, 2011
Engineered CD19 CAR-T cells showed dramatic complete remission in refractory B-ALL, marking a new era in adoptive immunotherapy.
NK Cell Therapy Innovation: Dr. Jeffrey Miller, University of Minnesota, 2005–Present
Showed feasibility and success of donor-derived NK cell infusions in treating leukemia.
iPSC-Derived Immune Cells for ALL: Dr. Shin Kaneko, Kyoto University, 2017
Pioneered generation of CAR-T and NK cells from induced pluripotent stem cells (iPSCs), potentially revolutionizing off-the-shelf immunotherapies [8-10].
Our clinical approach employs precision-based delivery strategies that amplify immune efficacy while reducing systemic toxicity:
Intravenous Infusion of CAR-T and NK Cells: Standard delivery method allowing broad systemic circulation, targeting circulating and marrow-resident leukemic blasts.
Intra-Bone Marrow Infusion: For direct engagement of leukemic microenvironments, particularly in sanctuary sites resistant to peripheral therapy.
Lymph Node-Targeted Immunotherapy: Emerging delivery system to restore antigen presentation in lymphoid-rich tissues.
Supportive MSC Co-Infusion: Used to mitigate therapy-related cytokine storms and GVHD in the setting of allogeneic infusions.
These delivery systems enhance both precision and durability of response in Cellular Immunotherapy for ALL [8-10].
At DrStemCellsThailand (DRSCT), we uphold rigorous ethical standards in sourcing and deploying stem cell therapies:
Wharton’s Jelly MSCs: Sourced from medically discarded umbilical cords, rich in immunomodulatory properties and ethically non-controversial.
Induced Pluripotent Stem Cells (iPSCs): Patient-specific cells reprogrammed and redifferentiated into personalized immune effector cells without embryo involvement.
Cord Blood-Derived Cells: Voluntarily donated and cryopreserved, these provide high compatibility and minimal ethical concerns.
GMP-Grade Expansion and Engineering: All cellular products undergo Good Manufacturing Practice (GMP)-compliant processing to ensure safety, traceability, and sterility.
By maintaining these high ethical standards, we advance Cellular Immunotherapies for Acute Lymphoblastic Leukemia (ALL) with both scientific integrity and humanitarian responsibility [8-10].
Preventing relapse in Acute Lymphoblastic Leukemia (ALL) hinges on timely immunological reprogramming and durable antileukemic surveillance. Our advanced immunotherapy protocols integrate:
By proactively deploying Cellular Immunotherapies for Acute Lymphoblastic Leukemia (ALL), we offer a dynamic, immune-mediated strategy to eradicate leukemic clones and fortify long-term remission stability [11-13].
Our hematology-oncology and cellular immunotherapy teams emphasize early intervention to maximize remission durability in ALL. Administering cellular therapies during minimal residual disease (MRD) phases or post-remission consolidation significantly improves survival metrics:
Patients treated early demonstrate superior progression-free survival (PFS), reduced relapse rates, and longer leukemia-free intervals. Our approach integrates early disease recognition with immunotherapy deployment to optimize outcomes in ALL [11-13].
ALL is driven by uncontrolled lymphoblast proliferation and immune evasion. Our Cellular Immunotherapies for Acute Lymphoblastic Leukemia (ALL) targets the disease at a molecular and immunologic level:
Our immunotherapy platform is tailored to dismantle ALL’s complex immunoevasive architecture while promoting lasting immunological control [11-13].
ALL progression spans a spectrum of immunologic derangements and leukemic burden. Cellular immunotherapies are adapted at each stage:
Stage 1: Pre-Leukemic State (Clonal Hematopoiesis or Germline Predisposition)
Stage 2: Overt ALL Diagnosis (Initial Lymphoblast Expansion)
Stage 3: Induction Remission Phase (Chemosensitive or Partial Response)
Stage 4: Minimal Residual Disease (MRD+)
Stage 5: Relapsed/Refractory Disease
Stage 1: Pre-Leukemic State
Stage 2: Newly Diagnosed ALL
Stage 3: Post-Induction/MRD+ Disease
Stage 4: Late MRD or Post-Transplant MRD
Stage 5: Relapsed/Refractory ALL
Our Cellular Immunotherapies for Acute Lymphoblastic Leukemia (ALL) program integrates:
Through cutting-edge immunoengineering and personalized delivery, we are redefining ALL treatment—moving from cytotoxic remission induction to immune-mediated, relapse-free survivorship [11-13].
By leveraging allogeneic cellular platforms, we offer rapid, effective, and scalable immunotherapeutic solutions for ALL, particularly for relapsed or transplant-ineligible populations [11-13].
Preventing relapse in Acute Lymphoblastic Leukemia (ALL) hinges on timely immunological reprogramming and durable antileukemic surveillance. Our advanced immunotherapy protocols integrate:
By proactively deploying Cellular Immunotherapies for Acute Lymphoblastic Leukemia (ALL), we offer a dynamic, immune-mediated strategy to eradicate leukemic clones and fortify long-term remission stability [14-16].
Our hematology-oncology and cellular immunotherapy teams emphasize early intervention to maximize remission durability in ALL. Administering cellular therapies during minimal residual disease (MRD) phases or post-remission consolidation significantly improves survival metrics:
Patients treated early demonstrate superior progression-free survival (PFS), reduced relapse rates, and longer leukemia-free intervals. Our approach integrates early disease recognition with immunotherapy deployment to optimize outcomes in ALL [14-16].
ALL is driven by uncontrolled lymphoblast proliferation and immune evasion. Our cellular immunotherapy strategy targets the disease at a molecular and immunologic level:
Our immunotherapy platform is tailored to dismantle ALL’s complex immunoevasive architecture while promoting lasting immunological control [14-16].
ALL progression spans a spectrum of immunologic derangements and leukemic burden. Cellular immunotherapies are adapted at each stage:
Stage 1: Pre-Leukemic State (Clonal Hematopoiesis or Germline Predisposition)
Stage 2: Overt ALL Diagnosis (Initial Lymphoblast Expansion)
Stage 3: Induction Remission Phase (Chemosensitive or Partial Response)
Stage 4: Minimal Residual Disease (MRD+)
Stage 5: Relapsed/Refractory Disease
Stage 1: Pre-Leukemic State
Stage 2: Newly Diagnosed ALL
Stage 3: Post-Induction/MRD+ Disease
Stage 4: Late MRD or Post-Transplant MRD
Stage 5: Relapsed/Refractory ALL
Our Cellular Immunotherapies for Acute Lymphoblastic Leukemia (ALL) program integrates:
Through cutting-edge immunoengineering and personalized delivery, we are redefining ALL treatment—moving from cytotoxic remission induction to immune-mediated, relapse-free survivorship [14-16].
By leveraging allogeneic cellular platforms, we offer rapid, effective, and scalable immunotherapeutic solutions for ALL, particularly for relapsed or transplant-ineligible populations [14-16].
Precision in cellular immunotherapy begins with molecular and immunophenotypic profiling of ALL subtypes. Each patient’s leukemia expresses a distinct surface antigen repertoire and genetic signature, guiding our tailored immunotherapeutic approach:
This strategy transforms ALL therapy into a dynamic, evolving immunologic dialogue, not a static protocol—empowering durable control through real-time immunologic adaptation [14-16].
Traditional ALL therapy—dominated by cytotoxic chemotherapy and transplantation—is being redefined by cellular immunotherapeutics that offer:
As ALL outcomes transition from chemotherapy-mediated remission to immune-mediated cure, cellular therapies are emerging as the new gold standard, particularly for high-risk and relapsed disease.
Post-cellular therapy monitoring integrates immune reconstitution, clonal dynamics, and molecular residual disease:
Through integrated biomarker platforms, we continuously evaluate immune efficacy and disease evolution, enabling precise response modulation.
Despite revolutionary success, cellular immunotherapies face barriers requiring innovation:
We are committed to advancing the field through next-generation designs, real-time analytics, and modular immunologic engineering [14-16].
Feature | Chemotherapy | Allogeneic HSCT | Cellular Immunotherapy |
---|---|---|---|
Target Specificity | Low | Moderate | High (antigen-specific) |
Toxicity Profile | High systemic toxicity | GVHD risk | CRS/ICANS (manageable) |
Durability of Remission | Moderate | High (with GVHD risk) | High (memory T cell-mediated) |
Relapse Rate in High-Risk ALL | >50% | ~30% | <20% post-CAR-T |
Treatment Timeframe | Months to years | Months | 2–4 weeks (infusion to response) |
Eligibility | Broad | Limited (age/comorbidities) | Expanding (including R/R and elderly) |
Cellular immunotherapy is redefining therapeutic value, patient eligibility, and survivorship potential in ALL [14-16].
These findings validate cellular immunotherapies as potent frontline or salvage strategies, with superior efficacy in relapsed, refractory, and MRD+ settings [14-16].
We are pioneering a pipeline of next-generation Cellular Immunotherapies for Acute Lymphoblastic Leukemia (ALL) technologies:
Our innovation strategy is anchored in durability, precision, safety, and global accessibility [14-16].
We recognize the disparities in access to cellular therapies, particularly in low- and middle-income countries. Our global agenda includes:
Through strategic partnerships and compassionate infrastructure, we aim to make cutting-edge ALL immunotherapy a global standard, not a localized luxury [14-16].
A detailed cost breakdown for Cellular Immunotherapies for Acute Lymphoblastic Leukemia (ALL) ranges from $25,000 to $75,000, depending on the complexity of the protocol, the type of cellular therapy utilized, and additional supportive interventions required. This pricing ensures accessibility to the most advanced and personalized immunotherapeutic treatments available