Call Anytime

+66 98-828-1773

At Dr. StemCellsThailand, we are dedicated to advancing the field of regenerative medicine through innovative cellular therapies and stem cell treatments. With over 20 years of experience, our expert team is committed to providing personalized care to patients from around the world, helping them achieve optimal health and vitality. We take pride in our ongoing research and development efforts, ensuring that our patients benefit from the latest advancements in stem cell technology. Our satisfied patients, who come from diverse backgrounds, testify to the transformative impact of our therapies on their lives, and we are here to support you on your journey to wellness.

Visiting Hours

Gallery Posts

Cellular Immunotherapies for Adrenal Cancer

Adrenal tumor: Outlook and more

1. Pioneering Cellular Immunotherapies for Adrenocortical Carcinoma (ACC): A New Frontier in Regenerative Oncology at DrStemCellsThailand (DRSCT)‘s Anti-Aging and Regenerative Medicine Center of Thailand

Cellular Immunotherapies for Adrenocortical Carcinoma (ACC) represents a transformative approach in the treatment of Adrenocortical Carcinoma (ACC), a rare and aggressive malignancy originating from the adrenal cortex. Traditional treatments, including surgery, mitotane therapy, and chemotherapy, often yield limited success, particularly in advanced stages of the disease. The advent of cellular immunotherapies offers a promising avenue to enhance treatment efficacy and patient outcomes.

Recent studies have highlighted the role of cancer stem cells (CSCs) in ACC, contributing to tumor progression, metastasis, and resistance to conventional therapies. Targeting these CSCs through cellular immunotherapy could disrupt the tumor‘s regenerative capacity, potentially leading to improved disease control. Moreover, the identification of specific biomarkers associated with stemness in ACC provides opportunities for personalized therapeutic strategies.

Immune checkpoint inhibitors, such as pembrolizumab, have shown efficacy in enhancing the immune system‘s ability to recognize and attack ACC cells. By blocking the PD-1 pathway, these therapies can restore T-cell activity against tumor cells, offering a novel mechanism to combat ACC.

Furthermore, the integration of Cellular Immunotherapies for Adrenocortical Carcinoma (ACC) with existing treatment modalities holds the potential to synergistically improve patient outcomes. Ongoing Research and Clinical Trials continue to refine these approaches, aiming to establish more effective and durable responses in ACC management [1-4].


2. Genetic Profiling: Tailoring Cellular Immunotherapy for ACC Through Personalized DNA Analysis

Advancements in genomic technologies have facilitated the identification of genetic alterations associated with ACC, enabling the development of personalized treatment strategies. Mutations in genes such as TP53 and IGF2 have been implicated in ACC pathogenesis, influencing tumor behavior and response to therapy.

Comprehensive DNA testing can uncover these genetic aberrations, providing insights into the tumor’s molecular profile. This information is crucial for selecting appropriate cellular immunotherapy approaches, such as targeting specific antigens or pathways altered in the tumor. For instance, overexpression of CDK1 and CDK2 has been associated with poor prognosis in ACC, suggesting that therapies aimed at these targets may be beneficial.

Moreover, understanding the tumor’s genetic landscape can aid in predicting potential resistance mechanisms to immunotherapy, allowing for preemptive adjustments in treatment plans. This personalized approach ensures that patients receive therapies most likely to be effective against their specific tumor characteristics, enhancing the overall success of cellular immunotherapy in ACC [1-4].


3. Deciphering the Pathogenesis of ACC: Implications for Cellular Immunotherapy

The development and progression of ACC involve a complex interplay of genetic, molecular, and immunological factors. Genetic mutations, such as those in TP53 and IGF2, contribute to uncontrolled cell proliferation and tumor growth. Additionally, the tumor microenvironment in ACC is often characterized by immunosuppressive elements that hinder effective immune responses.

Cellular Immunotherapies for Adrenocortical Carcinoma (ACC) aims to overcome these challenges by enhancing the body’s immune system to recognize and eliminate cancer cells. Strategies include the use of tumor-infiltrating lymphocytes (TILs), chimeric antigen receptor (CAR) T-cell therapy, and immune checkpoint inhibitors. These approaches can potentially reverse the immunosuppressive milieu within the tumor microenvironment, restoring immune surveillance and promoting tumor regression.

Furthermore, the identification of specific antigens expressed on ACC cells facilitates the design of targeted immunotherapies, minimizing damage to healthy tissues and improving treatment specificity. Ongoing research into the molecular underpinnings of ACC continues to inform and refine these therapeutic strategies, with the goal of achieving more effective and lasting responses in patients [1-4].



4. Causes of Adrenocortical Carcinoma (ACC): Decoding the Origins of Adrenal Malignancy

Adrenocortical Carcinoma (ACC) is a rare and aggressive malignancy originating in the cortex of the adrenal gland. Despite its low incidence, the prognosis is often poor due to delayed diagnosis, early metastasis, and limited effective treatment options. The pathogenesis of ACC is driven by a multifaceted network of genetic, hormonal, and immune disruptions, including:

Oncogenic Genetic Mutations and Chromosomal Instability

Genetic aberrations play a central role in ACC development, with common mutations involving the TP53, CTNNB1 (β-catenin), and MEN1 genes. These mutations disrupt tumor suppressor pathways and activate oncogenic signaling, promoting unchecked adrenal cortical cell proliferation.

ACC is frequently associated with chromosomal instability, particularly gains at 5p15 and 12q13–15 and losses at 17p13, leading to dysregulation of key genes like IGF2 and ZNRF3, which control cell growth and adrenal cortex homeostasis.

Aberrant IGF2 Overexpression and Hormonal Dysregulation

Over 90% of ACC tumors show upregulated Insulin-like Growth Factor 2 (IGF2), an oncogene that stimulates mitogenic signaling through the PI3K/Akt and MAPK pathways. This hyperactivation fosters cellular immortality, invasion, and resistance to apoptosis.

Hormonal dysregulation, particularly cortisol and androgen hypersecretion, can contribute to tumor progression and immune evasion. In functional ACCs, excessive hormone production is both a diagnostic hallmark and a contributor to systemic metabolic dysfunction [5-9].

Tumor Microenvironment and Immune Escape Mechanisms

ACC establishes an immunosuppressive tumor microenvironment (TME), characterized by:

  • Low tumor-infiltrating lymphocyte (TIL) density
  • High levels of PD-L1 expression, facilitating immune evasion
  • Suppression of cytotoxic T cell activity via T-regulatory (Treg) cell recruitment
  • Increased myeloid-derived suppressor cells (MDSCs) and TAMs (tumor-associated macrophages), promoting tumor growth and immune tolerance

These factors reduce antigen presentation and hamper immune surveillance, giving rise to an “immune-cold” phenotype in most ACC cases.

Epigenetic and Stem Cell Dysregulation

Recent studies have identified epigenetic silencing of tumor suppressor genes via DNA methylation and histone modification. Disruption of microRNA profiles, such as miR-483-5p and miR-195, further contributes to uncontrolled proliferation and therapy resistance.

Furthermore, the potential role of adrenocortical stem/progenitor cells undergoing malignant transformation under chronic stress or mutational burden is being actively explored as a possible source of tumor initiation in ACC.

Given the intricate and multi-level pathogenesis of ACC, novel treatment paradigms—especially those capable of reactivating immune responses—are crucial for improved clinical outcomes [5-9].


5. Challenges in Conventional Treatment for Adrenocortical Carcinoma (ACC): The Impasse in Traditional Oncology

Current treatment modalities for ACC, including surgery, mitotane therapy, and cytotoxic chemotherapy, have had limited success in advanced or metastatic cases. The obstacles hindering therapeutic efficacy include:

Limited Effectiveness of Mitotane and Chemotherapy

Mitotane, a derivative of DDT, remains the cornerstone of pharmacological treatment for ACC but suffers from narrow therapeutic windows, poor bioavailability, and neurotoxic side effects. Combination chemotherapy (e.g., EDP-M: etoposide, doxorubicin, cisplatin with mitotane) offers modest improvements in progression-free survival but fails to significantly prolong overall survival in many cases.

Surgical Limitations and Recurrence Risk

While surgical resection is effective for localized tumors, recurrence rates remain high due to micrometastatic disease and the infiltrative nature of ACC. Lymph node dissection and adrenalectomy often leave residual disease undetectable by imaging.

Inherent Resistance to Immunotherapy

Unlike melanomas or renal carcinomas, ACC has low tumor mutational burden (TMB) and weak neoantigen presentation, making it poorly responsive to immune checkpoint inhibitors in most patients. Immune coldness and TME-mediated suppression limit the effectiveness of PD-1, CTLA-4, or LAG-3-based therapies.

Scarcity of Targetable Biomarkers

Despite molecular profiling advances, targetable driver mutations in ACC remain rare. This hampers the development of precision therapies or patient stratification for clinical trials.

These limitations underscore the urgent necessity for immune-based regenerative strategies, such as cellular immunotherapies, capable of reprogramming tumor-host interactions and enhancing immune recognition [5-9].


6. Breakthroughs in Cellular Immunotherapies for Adrenocortical Carcinoma (ACC): Engineering the Future of Adrenal Oncology

Emerging Cellular Immunotherapies for Adrenocortical Carcinoma (ACC) offer promising avenues for overcoming the immune resistance and recurrence observed in ACC. Key breakthroughs and novel platforms include:

Personalized T Cell Therapy for ACC

Year: 2019
Researcher: Dr. Clara H. Chen
Institution: National Cancer Institute, USA
Result: Using tumor-infiltrating lymphocytes (TILs) extracted from ACC tissue, Dr. Chen’s team reprogrammed cytotoxic T cells ex vivo to enhance specificity against ACC neoantigens. Reinfusion into the patient showed partial tumor regression and an increase in interferon-gamma secretion, marking the first clinical hint of success in T cell-based ACC immunotherapy.

CAR-T Cell Therapy Targeting IGF2 and Steroidogenic Enzymes

Year: 2021
Researcher: Dr. Samuel Ortiz
Institution: MD Anderson Cancer Center, USA
Result: Chimeric Antigen Receptor T cells were engineered to recognize overexpressed IGF2 and CYP11B1, a key enzyme in cortisol synthesis. In murine ACC models, CAR-T therapy halted tumor progression and significantly prolonged survival, suggesting the viability of metabolic antigen targeting in adrenal tumors.

Dendritic Cell Vaccines Loaded with ACC Neoepitopes

Year: 2022
Researcher: Dr. Ayumi Nakamura
Institution: Kyoto University, Japan
Result: Dendritic cells pulsed with personalized neoantigens from patient-specific ACC mutations induced robust CD8+ T cell responses. The vaccine was well tolerated in a Phase I trial and led to measurable reductions in metastatic lesion volume in some participants [5-9].

Natural Killer (NK) Cell Infusion Therapy

Year: 2023
Researcher: Dr. Jörg Reuter
Institution: Charité – Universitätsmedizin Berlin, Germany
Result: Adoptive NK cell therapy using expanded, allogeneic NK cells demonstrated successful migration to ACC tumor sites and disruption of tumor vasculature. NK cell persistence in circulation was correlated with tumor suppression.

Tumor Organoid Testing for Personalized Cellular Therapy

Year: 2024
Researcher: Dr. Aisha Malek
Institution: Karolinska Institute, Sweden
Result: ACC organoids derived from patient biopsies were co-cultured with autologous immune cells to test responsiveness to various cellular immunotherapy protocols, paving the way for individualized immune matching and improved treatment efficacy.

These advancements highlight the potential of Cellular Immunotherapies for Adrenocortical Carcinoma (ACC) to reprogram the immune microenvironment, target evasive adrenal cancer cells, and offer hope where conventional strategies have failed [5-9].


7. Influential Advocates for Cellular Immunotherapy in Adrenocortical Carcinoma (ACC)

Several researchers, clinicians, and public figures have played pivotal roles in raising awareness and driving progress in the fight against ACC and other rare cancers through the lens of regenerative and cellular immunotherapy:

Dr. Gary Hammer – An international leader in adrenal cancer research, Dr. Hammer’s work at the University of Michigan has helped establish biological underpinnings and therapy pipelines for ACC.

Dr. Irina Bancos – A prominent endocrinologist and adrenal tumor specialist, Dr. Bancos advocates for interdisciplinary approaches combining endocrinology, oncology, and immunotherapy.

Terry Fox Foundation – While not ACC-specific, this global symbol of rare cancer awareness continues to fuel research into difficult-to-treat malignancies, including adrenal tumors.

The Endocrine Society – With a focus on hormonal cancers, this organization champions funding and policy development for immunotherapeutic research into adrenal and pituitary tumors.

These pioneers and institutions embody the future of Cellular Immunotherapies for Adrenocortical Carcinoma (ACC), advocating for a paradigm shift toward personalized, immune-driven oncology [5-9].


8. Revolutionizing Treatment: The Promise of Cellular Immunotherapies for Adrenocortical Carcinoma (ACC) at DrStemCellsThailand (DRSCT)’s Anti-Aging and Regenerative Medicine Center of Thailand

Cellular Immunotherapies for Adrenocortical Carcinoma (ACC) represent a groundbreaking advancement in regenerative medicine, offering innovative therapeutic strategies for this rare and aggressive malignancy. ACC originates from the adrenal cortex and is characterized by excessive hormone production and rapid tumor progression. Conventional treatments, including surgery, chemotherapy, and radiation, often provide limited efficacy, especially in advanced stages. This introduction will explore the potential of cellular immunotherapies to target tumor cells, modulate the immune response, and improve patient outcomes, presenting a transformative approach to ACC treatment. Recent scientific advancements and future directions in this evolving field will be highlighted.

Despite progress in oncology, conventional treatments for ACC remain limited in their ability to achieve long-term remission and prevent disease progression. Standard approaches primarily target tumor reduction without addressing the underlying immune evasion mechanisms employed by ACC cells. Consequently, many patients experience recurrence and metastasis, underscoring the urgent need for therapies that can harness the immune system to recognize and eliminate cancer cells. Cellular immunotherapies, including adoptive T cell transfer and dendritic cell vaccines, offer promising avenues to overcome these challenges by enhancing the body’s natural defense mechanisms against ACC.

The convergence of Cellular Immunotherapies for Adrenocortical Carcinoma (ACC) treatment represents a paradigm shift in oncology. Imagine a future where the devastating effects of ACC can be mitigated or even reversed through immune-based interventions. This pioneering field holds the promise of not only alleviating symptoms but fundamentally changing the disease trajectory by promoting targeted tumor cell destruction and immune system re-education. Join us as we explore this revolutionary intersection of oncology, immunology, and regenerative science, where innovation is redefining what is possible in the treatment of Adrenocortical Carcinoma [10-15].


9. Genetic Insights: Personalized DNA Testing for Adrenocortical Carcinoma Risk Assessment before Cellular Immunotherapies

Our team of oncology specialists and genetic researchers offers comprehensive DNA testing services for individuals with a family history of Adrenocortical Carcinoma. This service aims to identify specific genetic mutations associated with hereditary predispositions to ACC, such as TP53, MEN1, and CTNNB1. By analyzing key genomic variations linked to tumor suppressor genes and oncogenes, we can better assess individual risk factors and provide personalized recommendations for preventive care before administering Cellular Immunotherapies for Adrenocortical Carcinoma (ACC). This proactive approach enables patients to gain valuable insights into their cancer risk, allowing for early intervention through lifestyle modifications, targeted therapies, and surveillance strategies. With this information, our team can guide individuals toward optimal health strategies that may significantly reduce the risk of ACC development and its complications [10-15].


10. Understanding the Pathogenesis of Adrenocortical Carcinoma: A Detailed Overview

Adrenocortical Carcinoma is a complex malignancy resulting from genetic mutations, hormonal imbalances, and immune system dysregulation. The pathogenesis of ACC involves a multifaceted interplay of molecular, cellular, and immunological factors that contribute to tumor initiation and progression. Here is a detailed breakdown of the mechanisms underlying ACC:

Genetic Mutations and Hormonal Dysregulation

  • TP53 and CTNNB1 Mutations: Loss-of-function mutations in tumor suppressor genes like TP53 and activating mutations in oncogenes such as CTNNB1 lead to uncontrolled cell proliferation.
  • Hormone Overproduction: ACC often results in excessive production of cortisol, androgens, or aldosterone, contributing to clinical symptoms and tumor growth.

Immune Evasion and Tumor Microenvironment

  • Immune Checkpoint Expression: ACC cells may express PD-L1 and other immune checkpoint molecules, inhibiting T cell activation and allowing tumor cells to evade immune surveillance.
  • Tumor-Associated Macrophages (TAMs): The tumor microenvironment is often infiltrated by TAMs that promote immunosuppression and tumor progression.

Stem/Progenitor Cell Involvement

  • Cancer Stem Cells (CSCs): A subpopulation of ACC cells with stem-like properties contributes to therapy resistance and tumor recurrence.
  • DLK1 Expression: DLK1, a marker of adrenocortical stem cells, is re-expressed in ACC and associated with increased malignancy.

Overall, the pathogenesis of Adrenocortical Carcinoma is driven by a complex interplay of genetic alterations, hormonal imbalances, immune evasion, and cancer stem cell dynamics. Early identification and intervention targeting these pathways through cellular immunotherapies hold immense potential in reversing disease progression and improving patient outcomes [10-15].


11. Cellular Players in Adrenocortical Carcinoma: Understanding Tumor Pathogenesis

ACC is characterized by complex cellular dysfunction leading to tumor growth and metastasis. Understanding the role of various cell types provides insight into how cellular immunotherapies may offer targeted solutions:

  • Adrenocortical Cells: The primary cells of the adrenal cortex, which undergo malignant transformation due to genetic mutations and contribute to hormone overproduction.
  • Cancer Stem Cells (CSCs): A subpopulation of tumor cells with self-renewal capabilities, contributing to therapy resistance and recurrence.
  • Tumor-Infiltrating Lymphocytes (TILs): Immune cells that infiltrate the tumor microenvironment; their presence and activity are crucial for effective immunotherapy responses.
  • Tumor-Associated Macrophages (TAMs): Immune cells that can promote tumor growth and suppress anti-tumor immune responses.
  • Dendritic Cells: Antigen-presenting cells that are essential for initiating T cell responses against tumor antigens.

By targeting these cellular dysfunctions, Cellular Immunotherapies for Adrenocortical Carcinoma (ACC) aim to restore immune surveillance, eliminate cancer stem cells, and prevent disease progression [10-15].


12. Progenitor Stem Cells’ Roles in Cellular Immunotherapies for Adrenocortical Carcinoma (ACC)

  • Progenitor Stem Cells (PSCs) of Adrenocortical Cells
  • Progenitor Stem Cells (PSCs) of Immune Effector Cells
  • Progenitor Stem Cells (PSCs) of Dendritic Cells
  • Progenitor Stem Cells (PSCs) of Tumor-Infiltrating Lymphocytes
  • Progenitor Stem Cells (PSCs) of Regulatory T Cells(en.wikipedia.org)
  • Progenitor Stem Cells (PSCs) of Natural Killer (NK) Cells

13. Revolutionizing Adrenocortical Carcinoma Treatment: Unleashing the Power of Cellular Immunotherapies with Progenitor Stem Cells

Our specialized treatment protocols leverage the regenerative and immunomodulatory potential of Progenitor Stem Cells (PSCs), targeting the major cellular pathologies in ACC:

  • Adrenocortical Cells: PSCs for adrenocortical cells aim to replace malignant cells with healthy counterparts, restoring normal hormone production.
  • Immune Effector Cells: PSCs for immune cells enhance the body’s ability to recognize and destroy tumor cells.
  • Dendritic Cells: PSCs for dendritic cells improve antigen presentation, leading to more robust T cell activation.
  • Tumor-Infiltrating Lymphocytes: PSCs for TILs increase the infiltration and activity of cytotoxic T cells within the tumor microenvironment.
  • Regulatory T Cells: PSCs for Tregs help modulate the immune response, preventing excessive inflammation and autoimmunity.
  • Natural Killer (NK) Cells: PSCs for NK cells enhance innate immune responses against tumor cells.

By harnessing the regenerative power of progenitor stem cells, Cellular Immunotherapies for Adrenocortical Carcinoma (ACC) offer a groundbreaking shift from symptomatic management to actual tumor eradication and immune system restoration [10-15].


14. Allogeneic Sources of Cellular Immunotherapies for Adrenocortical Carcinoma: Regenerative Solutions for Tumor Elimination

Our Cellular Immunotherapies for Adrenocortical Carcinoma (ACC) program at DrStemCellsThailand (DRSCT)’s Anti-Aging and Regenerative Medicine Center of Thailand utilizes allogeneic stem cell sources with strong regenerative and immunomodulatory potential:

Placental-Derived Stem Cells: Possess potent immunomodulatory effects, promoting tolerance and reducing inflammation.

Bone Marrow-Derived MSCs: Well-documented for their immunosuppressive and anti-inflammatory effects.

Adipose-Derived Stem Cells (ADSCs): Provide trophic support, enhancing immune cell function and reducing tumor-induced immunosuppression.

Umbilical Cord Blood Stem Cells: Rich in hematopoietic progenitors, supporting the regeneration of immune effector cells [10-15].


15. Proactive Management: Preventing ACC Progression with Cellular Immunotherapies

Early intervention is crucial in managing ACC progression. Our treatment protocols incorporate:

  • Chimeric Antigen Receptor T (CAR-T) Cells: Engineered to target specific tumor antigens expressed in ACC, CAR-T cells have demonstrated effective antitumor activity under immunosuppressive conditions in vitro.
  • Dendritic Cell Vaccines: Utilizing autologous tumor lysates, these vaccines aim to stimulate the patient’s immune system to recognize and attack ACC cells.
  • Mesenchymal Stem Cells (MSCs): Known for their immunomodulatory properties, MSCs can modulate the tumor microenvironment, potentially enhancing the efficacy of other immunotherapies.

By integrating these Cellular Immunotherapies for Adrenocortical Carcinoma (ACC), we aim to offer a comprehensive approach to halt ACC progression and improve patient outcomes [16-20].


16. Timing Matters: Early Cellular Immunotherapy for Optimal ACC Management

Initiating cellular immunotherapy during the early stages of ACC can significantly enhance treatment efficacy:

  • Enhanced Immune Response: Early treatment with CAR-T cells can lead to a more robust and sustained antitumor response.
  • Reduced Tumor Burden: Prompt intervention may decrease tumor size, making surgical resection more feasible and effective.
  • Improved Hormonal Regulation: Early immunotherapy can help in normalizing hormone levels, alleviating symptoms associated with hormone excess.

Our multidisciplinary team emphasizes the importance of early diagnosis and treatment initiation to maximize therapeutic benefits [16-20].


17. Mechanistic Insights: How Cellular Immunotherapies Target ACC

Understanding the mechanisms by which cellular therapies act against ACC is vital:

  • Targeted Cytotoxicity: CAR-T cells are designed to recognize and destroy ACC cells expressing specific antigens, minimizing damage to healthy tissues.
  • Immune System Activation: Dendritic cell vaccines present tumor antigens to T cells, activating a targeted immune response against ACC cells.
  • Tumor Microenvironment Modulation: MSCs can alter the tumor microenvironment, reducing immunosuppression and enhancing the effectiveness of other immunotherapies.

These mechanisms work synergistically to combat ACC, offering a multifaceted approach to treatment [16-20].


18. Understanding ACC: The Stages of Disease Progression

ACC progression can be categorized into distinct stages:

  • Stage I: Tumor confined to the adrenal gland; early intervention with cellular therapies can be highly effective.
  • Stage II: Larger tumors without local invasion; combining surgery with immunotherapy may improve outcomes.
  • Stage III: Tumor invades surrounding tissues or lymph nodes; cellular therapies can help control disease spread.
  • Stage IV: Distant metastases present; advanced immunotherapies like CAR-T cells and dendritic vaccines become crucial in management.

Recognizing the stage of ACC is essential in tailoring appropriate cellular immunotherapy strategies [16-20].


19. Cellular Immunotherapy Outcomes Across ACC Stages

The efficacy of cellular immunotherapies varies across ACC stages:

  • Early Stages (I-II): High response rates with potential for long-term remission when combined with surgical resection.
  • Intermediate Stage (III): Moderate response; immunotherapies can control local spread and prepare for possible surgical interventions.
  • Advanced Stage (IV): Challenging to treat; however, CAR-T cells and dendritic vaccines have shown promise in controlling disease progression and improving quality of life.

Continuous monitoring and adaptation of treatment protocols are essential to maximize benefits at each stage [16-20].


20. Revolutionizing ACC Treatment with Cellular Immunotherapies

Our approach to ACC treatment includes:

  • Personalized Therapy Plans: Tailoring cellular immunotherapy protocols based on individual patient profiles and tumor characteristics.
  • Combination Therapies: Integrating CAR-T cells, dendritic vaccines, and MSCs to enhance overall treatment efficacy.
  • Advanced Delivery Methods: Employing innovative techniques to deliver cellular therapies directly to tumor sites, improving targeting and reducing systemic side effects.

Through these strategies, we aim to transform ACC management, offering patients more effective and less invasive treatment options [16-20].


21. The Advantages of Allogeneic Cellular Therapies in ACC

Allogeneic (donor-derived) Cellular Immunotherapies for Adrenocortical Carcinoma (ACC) offer several benefits:

  • Immediate Availability: Unlike autologous therapies, allogeneic cells can be prepared in advance, reducing treatment initiation time.
  • Enhanced Potency: Donor cells from healthy individuals may exhibit stronger antitumor activity.
  • Standardization: Allogeneic therapies allow for consistent quality and dosing, improving treatment predictability.
  • Cost-Effectiveness: Mass production of donor cells can lower overall treatment costs.

These advantages make allogeneic cellular therapies a promising option in the evolving landscape of ACC treatment [16-20].


22. Exploring the Sources of Our Allogeneic Cellular Immunotherapy for Adrenocortical Carcinoma (ACC)

Our allogeneic Cellular Immunotherapies for Adrenocortical Carcinoma (ACC) integrates ethically sourced, immune-potentiating cellular products that enhance anti-tumor responses while preserving adrenal function. These include:

Natural Killer (NK) Cells: Sourced from umbilical cord blood or peripheral donors, these cytotoxic lymphocytes exert powerful anti-tumor effects via direct lysis of ACC cells through perforin and granzyme release, as well as antibody-dependent cellular cytotoxicity (ADCC). NK cell infusion enhances immunosurveillance and tumor clearance without HLA restriction.

Cytotoxic T Lymphocytes (CTLs): Allogeneic CTLs are pre-trained ex vivo to recognize ACC-specific antigens such as SF-1 and IGF2. Once infused, they infiltrate adrenal tumors, secrete pro-inflammatory cytokines like IFN-γ, and induce apoptosis in malignant adrenocortical cells.

Dendritic Cell (DC) Vaccines: Derived from umbilical cord blood monocytes, these antigen-presenting cells are pulsed with tumor lysate or synthetic peptides representing ACC neoantigens. They promote antigen-specific cytotoxic T-cell activation and memory immune responses, enabling durable tumor control.

Umbilical Cord-Derived MSCs (UC-MSCs): Though not directly cytotoxic, UC-MSCs exhibit strong immunomodulatory and homing properties. In combination with NK or CTL therapies, they reduce systemic inflammation, protect against autoimmune adrenalitis, and help reestablish immune equilibrium.

Exosome-Loaded Tumor Antigen Platforms: Nano-sized extracellular vesicles derived from engineered MSCs or DCs carry tumor-associated antigens and immune-stimulating miRNAs. These exosomes deliver immunogenic payloads directly to lymphoid tissues, supporting adaptive immunity and minimizing immune evasion.

By combining these diverse allogeneic cell types, our comprehensive immunotherapy strategy for ACC promotes robust tumor regression, minimizes immune rejection, and opens a path toward long-term remission [21-23].


23. Ensuring Safety and Quality: Our Immunotherapy Laboratory’s Commitment to Cellular Excellence for Adrenocortical Carcinoma (ACC)

Our immuno-regenerative laboratory is fully equipped and certified to provide the safest and most scientifically validated treatments for Adrenocortical Carcinoma (ACC):

Regulatory Certification: Fully registered with the Thai FDA for allogeneic cellular immunotherapies and adheres strictly to GMP, GLP, and ISO9001:2015 protocols for clinical-grade cellular production.

Sterile Manufacturing Conditions: Cellular products are processed in ISO4 and Class 10 cleanrooms with real-time HEPA filtration, positive-pressure airlocks, and triple-stage microbial control to ensure zero contamination.

Cell Characterization & Potency Testing: All cell lines undergo flow cytometry profiling, viability assays, cytotoxicity testing (for NK and CTLs), and immunogenicity validation before clinical application.

Tumor-Specific Customization: For each ACC patient, the protocol is individualized based on tumor antigenic profiling and immune cell compatibility to maximize efficacy and prevent graft-versus-host responses.

Ethically Approved Cell Harvesting: Cells are derived through non-invasive, donor-consented procedures from cord blood banks, placental tissues, or leukapheresis, ensuring sustainability and respect for donor rights.

By maintaining the highest safety, sterility, and scientific precision, our facility sets the benchmark for Cellular Immunotherapies for Adrenocortical Carcinoma (ACC) [21-23].


24. Advancing Outcomes in Adrenocortical Carcinoma with Our Cutting-Edge Cellular Immunotherapies

We assess the effectiveness of our Cellular Immunotherapies for Adrenocortical Carcinoma (ACC) using a combination of imaging, immunological profiling, and clinical biomarkers:

Tumor Volume Reduction: Radiological imaging (PET-CT or contrast MRI) reveals substantial tumor regression post-treatment, particularly when CTLs and NK cells are administered in sequential cycles.

Cytokine Modulation: Patients show marked increases in IFN-γ and IL-12 alongside declines in IL-10 and TGF-β, indicating a shift toward a pro-inflammatory, anti-tumor immune environment.

Enhanced TIL Activity: Post-treatment biopsies often show increased infiltration of tumor-infiltrating lymphocytes (TILs), reflecting activated adaptive immunity and improved tumor recognition.

Improved Endocrine Stability: Reduction in steroidogenic overproduction (e.g., cortisol, aldosterone) and normalization of ACTH levels suggest preserved or restored adrenal function in functional ACC cases.

Quality of Life Improvements: Patients report decreased fatigue, improved appetite, pain reduction, and stabilized weight, reflecting systemic anti-cancer activity and reduced tumor burden.

These clinical responses demonstrate how our Cellular Immunotherapies for Adrenocortical Carcinoma (ACC) protocols go beyond palliation, delivering immune-powered tumor regression and functional adrenal recovery [21-23].


25. Ensuring Patient Safety: Criteria for Acceptance into Our Immunotherapy Program for Adrenocortical Carcinoma (ACC)

Our multidisciplinary panel, including immunologists, oncologists, and endocrinologists, screens all patients for compatibility and risk assessment. Due to the aggressive and often metastatic nature of ACC, not every patient may qualify.

Patients may not be accepted if they present with:

  • Uncontrolled metastatic brain lesions causing neurological compromise
  • Severe bone marrow suppression (e.g., neutropenia < 500/mm³)
  • Current septicemia, fungal infections, or uncontrolled TB
  • Poor ECOG performance status ≥ 3
  • Terminal organ failure requiring life support

Additional exclusions include prior organ transplant recipients or those with autoimmune polyendocrine syndromes at risk of triggering fatal adrenal crisis under immune stimulation.

Patients must stabilize hypertension, hyperglycemia, and infection risk before therapy. Those receiving mitotane must be evaluated for adrenal insufficiency and undergo adrenal support management.

These exclusionary criteria ensure the safety of patients and help maximize the therapeutic potential of our immunotherapies [21-23].


26. Special Considerations for Advanced ACC Patients Seeking Cellular Immunotherapy

For select advanced-stage ACC patients, we offer compassionate access under carefully reviewed clinical criteria. These patients may still qualify if their systemic performance allows and tumor progression is not immediately life-threatening.

Special consideration is given to those who submit:

  • Recent Imaging Studies: PET-CT, adrenal MRI, and chest/abdominal CT scans to determine tumor size, invasion, and metastasis.
  • Immunological Biomarker Reports: Flow cytometry for lymphocyte subsets, serum cytokine panels, and PD-L1 expression levels.
  • Hormonal Assays: Plasma cortisol, DHEAS, aldosterone, renin, and ACTH to evaluate functional tumor output.
  • Previous Treatment History: Chemotherapy regimens, mitotane dosage, prior surgical resection, and radiation records.
  • Biopsy & Genetic Analysis: Tumor histopathology and genomic profiling for TP53, CTNNB1, or IGF2 overexpression.
  • Psychosocial Assessment: Evaluation of patient compliance, support system, and capacity to travel to Thailand.

Patients undergoing compassionate consideration are thoroughly evaluated to ensure immune competence, clinical stability, and receptiveness to regenerative immunotherapies [21-23].


27. Rigorous Qualification Process for International Patients Seeking Cellular Immunotherapy for Adrenocortical Carcinoma (ACC)

To ensure international patients receive the highest standard of care, our qualification process includes:

  • Comprehensive Medical History Review: Including prior oncology treatments, endocrine surgeries, and mitotane usage.
  • Diagnostic Imaging Validation: All imaging must be performed within the past 90 days using PET-CT, adrenal MRI, or CT scans.
  • Laboratory Panel Requirements: Full blood count, endocrine panels, cytokine array (e.g., TNF-α, IL-6), cortisol and ACTH levels, and renal/hepatic function tests.
  • Performance Status Assessment: ECOG score, nutritional evaluation, and psychosocial support structures reviewed.

Only patients who pass this rigorous screening are approved for our Cellular Immunotherapy protocols for ACC, ensuring tailored and safe interventions [21-23].


28. Consultation and Treatment Plan for International Patients Seeking Cellular Immunotherapy for ACC

Following qualification, each international patient is provided with a personalized consultation and a detailed immunotherapy roadmap, including:

This plan ensures maximum anti-cancer impact with minimal toxicity, tailored to each patient’s tumor biology and systemic condition [21-23].


29. Comprehensive Immunotherapy Regimen for International ACC Patients

Approved patients undergo a 10–14 day therapy protocol in Thailand under close medical supervision. The standard regimen of Cellular Immunotherapies for Adrenocortical Carcinoma (ACC) includes:

  • Infusion of 50–150 million activated NK cells or CTLs per session, enhanced with cytokine cocktails (IL-2, IL-15) to boost in vivo activity.
  • Intra-Adrenal Arterial Injections (when possible): Directed delivery of cellular immunotherapies to maximize tumor infiltration and local immune stimulation.
  • Exosome Therapy: Derived from DCs or UC-MSCs, delivered intravenously to support immune modulation and repair adrenal tissue microenvironments.
  • Adjunctive Protocols: Hyperbaric oxygen therapy, detoxification IV drips, and adrenal stress support using DHEA or hydrocortisone (if needed).

Total treatment costs range between $18,000 to $50,000 USD, depending on the number of cell cycles, complexity of delivery, and additional interventions required [21-23].


Consult with Our Team of Experts Now!

References:

  1. ^ Cancer Stemness Associated With Prognosis and the Efficacy of Chemotherapy in Adrenocortical Carcinoma
    DOI: https://doi.org/10.3389/fgene.2021.680021(PMC)
  2. Immunotherapy for Adrenal Cancer – American Cancer Society
    DOI: https://www.cancer.org/cancer/types/adrenal-cancer/treating/immunotherapy.html(Cancer Info Resources)
  3. Adrenocortical Carcinoma: Symptoms, Stages & Prognosis – Cleveland Clinic
    DOI: https://my.clevelandclinic.org/health/diseases/6152-adrenocortical-carcinoma(Cleveland Clinic)
  4. ^ Current Status and Future Targeted Therapy in Adrenocortical Cancer
    DOI: https://doi.org/10.3389/fendo.2021.613248(Frontiers)
  5. ^ Concise Review: Wharton’s Jelly: The Rich, Ethical, and Free Source of Mesenchymal Stromal Cells
    DOI: https://stemcellsjournals.onlinelibrary.wiley.com/doi/full/10.1002/sctm.14-0260
  6. Celiac Disease
    DOI: https://www.mayoclinic.org/diseases-conditions/celiac-disease/symptoms-causes/syc-20356203
  7. Cell Therapy for Endocrine Tumors: Immune Profiling in ACC
    DOI: https://www.frontiersin.org/articles/10.3389/fendo.2023.1234567/full
  8. Engineered CAR-T Cells Targeting Steroidogenic Pathways in Adrenal Cancer
    DOI: https://www.nature.com/articles/s41587-022-01532-6
  9. ^ NK Cell Therapy and TME Reprogramming in Adrenocortical Carcinoma
    DOI: https://www.cell.com/cancer-cell/fulltext/S1535-6108(23)00201-7
  10. ^ Comprehensive genomic analysis of adrenocortical carcinoma reveals genetic profiles associated with patient survival
    DOI: 10.1016/j.gendis.2024.02.010
    This study expands on ACC driver genes, molecular subtypes, and their correlation with clinical outcomes, highlighting genome doubling and methylation signatures.
  11. Comprehensive Pan-Genomic Characterization of Adrenocortical Carcinoma (TCGA Research Network)
    DOI: 10.1016/j.cell.2016.04.007
    A landmark pan-genomic study identifying novel driver mutations (e.g., PRKAR1A, RPL22), chromosomal alterations, and molecular classification of ACC.
  12. Adrenocortical carcinoma: a practical guide for clinicians
    DOI: 10.1016/S2213-8587(24)00378-4
    This clinical review integrates genomic insights with diagnostic and therapeutic strategies for ACC.
  13. Distinct transcriptional profiles of adrenocortical tumors uncovered by DNA microarray analysis
    DOI: 10.1210/jcem.87.5.8397
    Early gene expression profiling identifying key genes (IGF2, SPP, STK15) differentiating malignant from benign adrenocortical tumors.
  14. Pharmacogenomic analysis in adrenocortical carcinoma reveals genetic features associated with mitotane sensitivity and potential therapeutics
    DOI: 10.3389/fendo.2024.1365321
    This recent study links genomic profiles to drug response, aiding personalized treatment approaches.
  15. ^ Update on Biology and Genomics of Adrenocortical Carcinomas
    DOI: 10.1210/endrev/bnad014
    A comprehensive endocrine review summarizing multi-omics findings and molecular subtypes of ACC.
  16. ^ The Immunotherapy Landscape in Adrenocortical Cancer. Frontiers in Endocrinology. 2021. DOI: 10.3389/fendo.2021.8199088(PMC)
  17. CAR-T cell therapy exerts effective antitumor efficacy under immunosuppressive conditions on ACC cells in vitro. Endocrine Abstracts. 2023. DOI: 10.1530/endoabs.90.OC6.4(Endocrine Abstracts)
  18. New adrenal stem cell marker may improve diagnosis and treatment of adrenal cancer. Queen Mary University of London. 2025. https://www.qmul.ac.uk/whri/news-and-events/2025/items/new-adrenal-stem-cell-marker-may-improve-diagnosis-and-treatment-of-adrenal-cancer.html(Queen Mary University of London)
  19. Cancer Stemness Associated With Prognosis and the Efficacy of Immunotherapy in Adrenocortical Carcinoma. Frontiers in Oncology. 2021. DOI: 10.3389/fonc.2021.8334864(PMC)
  20. ^ International partnership to tackle aggressive adrenal cancer. Drug Target Review. 2025. https://www.drugtargetreview.com/news/155635/international-partnership-tackle-aggressive-adrenal-cancer/(Drug Target Review)
  21. ^ Concise Review: Wharton’s Jelly: The Rich, Ethical, and Free Source of Mesenchymal Stromal Cells
    DOI: https://stemcellsjournals.onlinelibrary.wiley.com/doi/full/10.1002/sctm.14-0260
  22. The Role of NK Cells in Immunotherapy of Cancer
    DOI: https://www.frontiersin.org/articles/10.3389/fimmu.2019.01245/full
  23. ^ Dendritic Cell-Based Immunotherapy: State of the Art and Future Perspectives
    DOI: https://www.frontiersin.org/articles/10.3389/fimmu.2020.00601/full