top of page

Graft-Versus-Host Disease: Complications and Prevention Methods


Graft-versus-host disease (GvHD) stands out as one of the prevalent complications associated with stem cell transplantation. This condition arises when immune cells from the transplanted graft recognize the recipient's body cells as foreign, leading to an immune response against the host's tissues. The term "graft" refers to the transplanted or donated tissue, and "host" refers to the tissues of the individual receiving the transplant. This condition is frequently encountered as a complication following allogeneic hematopoietic stem cell transplantation (HCT), where the immune cells in the transplanted graft react against the host's tissues. Additional materials on GvHD may include information on its classification, symptoms, risk factors, and potential treatment options. Moreover, exploring the impact of GvHD on various organs and tissues in the body could provide a comprehensive understanding of the disorder. Additionally, insights into ongoing research and advancements in managing GvHD could contribute to a more thorough overview of the topic.


The prevalence and impact of GVHD in the context of HCT depend on several factors, particularly the degree of human leukocyte antigen (HLA) matching between the donor and recipient. Acute GVHD is observed in approximately 50% of patients who undergo HCT from an HLA-matched sibling, suggesting a substantial risk even in situations where HLA compatibility is relatively high. However, the occurrence of acute GVHD tends to be even more frequent when donors are unmatched. The incidence of chronic GVHD exhibits a wide range, spanning from 6% to 80%. This variability underscores the complexity of GVHD dynamics and the influence of diverse factors in its development. Notably, GVHD is recognized as a significant contributor to morbidity and mortality following HCT, with over 10% of patients succumbing to complications associated with this condition. This underscores the critical importance of understanding and managing GVHD to enhance the overall success of HCT procedures.



Various risk factors contribute to the likelihood of developing acute GVHD. These include higher degrees of HLA mismatch, a history of prior acute GVHD, gender disparities between the donor and recipient, advanced age of either the donor or recipient, peripheral stem cell recipients, alloimmunization of the donor, and the seropositivity of the donor for Cytomegalovirus and Epstein Barr virus. This comprehensive list emphasizes the multifaceted nature of GVHD risk, incorporating genetic, immunological, and virological factors. Understanding these risk factors is crucial for identifying individuals at higher risk and implementing preventive measures or tailored therapeutic strategies to mitigate the impact of GVHD on transplant outcomes. As such, ongoing research in this field aims to refine risk prediction models and improve preemptive interventions to enhance the overall success and safety of HCT.


Understanding The Classification of GVHD

The classification of GVHD is traditionally based on the timing of its presentation following a HCT, with a defined cutoff of 100 days post-transplant. This temporal categorization has led to the distinction between acute and chronic GVHD. The National Institutes of Health (NIH) has further refined this classification by considering specific clinical manifestations.



  1. Acute Classic GVHD: Denotes the typical presentation of acute GVHD within the initial 100 days post-transplant. This phase involves damage to the skin, liver, and gastrointestinal tract, leading to symptoms like skin rash, jaundice, and diarrhea.

  2. Persistent, Recurrent, or Late-Onset Acute GVHD: "Persistent" indicates ongoing symptoms without resolution, "recurrent" signifies a reappearance after initial improvement, and "late-onset" points to acute GVHD occurring beyond the usual 100-day timeframe. These variations highlight the dynamic and sometimes prolonged nature of acute GVHD.

  3. Classic Chronic GVHD: This type typically manifests more than 100 days post-transplant. Chronic GVHD involves long-term complications affecting various organs, including the skin, eyes, mouth, liver, and lungs. Symptoms range from skin changes to joint pain, dry eyes, and oral issues, requiring a nuanced approach for effective management.

  4. Overlap Syndrome: In this type, features of both acute and chronic GVHD coexist or when distinguishing between the two becomes challenging.


By differentiating between acute and chronic phases and considering variations in onset and clinical features, the classification provides a framework for more accurate diagnosis, prognosis, and treatment strategies tailored to the specific manifestations of GVHD observed in a patient.


Understanding the Settings of GvHD

GvHD is a complex immune-mediated condition that can manifest in distinct medical scenarios, each involving unique transplantation settings. Understanding the contexts in which GvHD occurs is crucial for predicting, preventing, and managing this complication effectively.


1. Following Allogeneic Bone Transplantation (Most Common):

In allogeneic bone transplantation, where bone marrow or stem cells from a donor are transplanted into a recipient, GvHD is most commonly observed. This occurs when the transplanted immune cells from the donor recognize the recipient's tissues as foreign and mount an immune response, resulting in tissue damage.


2. Following Transplantation of Solid Organs Rich in Lymphoid Cells (e.g., Liver):

GvHD can also occur after the transplantation of solid organs that are rich in lymphoid cells, with the liver being a notable example. In such cases, the immune cells in the transplanted organ may perceive the recipient's tissues as foreign, leading to an immune reaction similar to that seen in bone transplantation.


3. Following Transfusion of Un-Irradiated Blood:

Transfusion-related GvHD is a rare but serious complication that can arise when blood from a donor contains viable T lymphocytes. If this un-irradiated blood is transfused into an immunocompromised recipient, the T cells in the donor blood can recognize the recipient's tissues as foreign, initiating an immune response and potentially causing GvHD.


In each of these settings, the common theme is the introduction of immunologically competent cells from a donor into an immunocompromised recipient. The key factor in GvHD development is the recognition of host tissues as foreign by the transplanted immune cells, triggering an immune response against the recipient's body. Managing and preventing GvHD in these scenarios often involves strategies to modulate the immune response, such as immunosuppressive medications, careful donor selection, and, in the case of transfusions, irradiation of blood products to eliminate T cells.


Understanding the Factors

To delve further into the factors influencing the occurrence and severity of GvHD, it is essential to understand the immunological dynamics and various components involved in the transplantation process. GvHD arises when immunologically competent cells from the donor are transplanted into recipients with compromised immune systems. These transferred cells, typically T lymphocytes, recognize alloantigens in the host tissues as foreign, initiating an immune response against the recipient's cells. The interaction between the donor's immune cells and the host's tissues plays a central role in the development of GvHD.


  1. Donor-Host Factors: The human leukocyte antigen (HLA) system, a critical determinant in this regard, is crucial for minimizing the risk of GvHD. However, even with a high degree of HLA matching, genetic differences in minor histocompatibility antigens can contribute to the immune response. Donor-recipient age and sex differences also play a role in influencing the risk of GvHD.

  2. Stem-Cell Source Factors: The choice of stem cell source for transplantation introduces another layer of complexity. Bone marrow, peripheral blood, and umbilical cord blood are common sources, each with distinct cellular compositions. For instance, peripheral blood grafts have a higher concentration of mature T cells, potentially increasing the risk of GvHD. Additionally, graft manipulation techniques, such as T-cell depletion or specific engineering approaches, can impact the immune cell composition and function, influencing GvHD outcomes.

  3. Immune Modulation Factors: Prophylactic use of immunosuppressive medications, such as calcineurin inhibitors and methotrexate, is standard practice to suppress donor T cell activity. The challenge lies in achieving a delicate balance between preventing GvHD and maintaining sufficient immune function to combat infections and tumors. Ongoing research explores advanced techniques, including targeted immunosuppression and regulatory T cell therapy, to enhance the precision and efficacy of immune modulation.

  4. Chemotherapy and Radiation Therapy: The conditioning regimen, comprising chemotherapy and/or radiation therapy, is designed to prepare the recipient for transplantation by suppressing their immune system. However, this regimen also induces tissue damage and inflammation, contributing to the complexity of GvHD development. The intensity, type, and duration of the conditioning regimen are crucial factors influencing the degree of immune suppression and subsequent GvHD risk.


Understanding The Pathogenesis Phases

The pathogenesis of GvHD involves a series of intricate immunological events driven primarily by the recognition of host tissues as foreign by donor cytotoxic CD8 T cells. This recognition leads to the proliferation of these CD8 T cells, resulting in a severe organ-damaging immune response characterized as a type IV cytotoxic T cell hypersensitivity reaction. The progression of GvHD can be delineated into three distinct phases:


  1. Phase 1 (Afferent Phase): Initiating the GvHD process, the afferent phase commences with the introduction of donor cells into the recipient's body during transplantation. In this initial stage, activated donor T cells discern the host's tissues as foreign, instigating the immune response. The afferent phase involves the activation and priming of immune cells, particularly T cells, which then migrate towards target tissues, setting the foundation for subsequent phases in the GvHD progression.

  2. Phase 2 (Efferent Phase): Advancing from the afferent phase, the efferent phase witnesses an escalation of the immune response, leading to the infiltration of activated immune cells into various organs and tissues. This phase is characterized by the dissemination of inflammatory signals, cytokines, and chemokines, contributing to tissue damage and inflammation. Organs like the skin, liver, and gastrointestinal tract are notably affected during the efferent phase. It signifies the amplification and systemic dissemination of the immune response, resulting in clinical manifestations and symptoms associated with GvHD.

  3. Phase 3 (Effector Phase): The effector phase represents the apex of GvHD progression, marked by the emergence of clinical symptoms and extensive tissue damage. In this phase, donor immune cells exhibit full-fledged effector functions, causing widespread tissue injury and dysfunction. Clinical manifestations may include skin issues like rash and blistering, liver complications leading to jaundice, and gastrointestinal symptoms such as diarrhea and abdominal pain. The effector phase underscores the substantial impact of GvHD on the overall health of the transplant recipient, necessitating swift and targeted interventions to mitigate the severity of the disease.

Understanding these distinct phases provides insights into the sequential events that contribute to the development and progression of GvHD. Targeting specific stages of this process may offer potential therapeutic opportunities to intervene and mitigate the severity of GvHD. Strategies aimed at modulating immune responses, controlling cytokine release, or inhibiting the migration of cytotoxic cells could be explored to enhance the safety and success of HCT.


Understanding The Affected Area

The manifestations of GvHD extend beyond systemic effects and often involve specific organ systems, each exhibiting distinct histopathological features. Understanding the tissue-level changes provides valuable insights into the clinical impact and severity of GvHD.


  1. Gastrointestinal Tract: GvHD often exerts a pronounced impact on the gastrointestinal tract, leading to a range of symptoms and histopathological changes. In the early stages, patients may experience diarrhea, abdominal pain, and gastrointestinal bleeding. The tissue-level changes involve inflammation, epithelial cell damage, and alterations in the mucosal lining. Severe cases can progress to gut barrier dysfunction, allowing the translocation of bacteria and other pathogens, further exacerbating the immune response.

  2. Liver: The liver is another major target of GvHD. Hepatic GvHD often presents with symptoms such as jaundice, hepatomegaly (enlarged liver), and abnormal liver function tests. At the tissue level, GvHD manifests as inflammation and damage to hepatic cells, potentially leading to fibrosis and impaired liver function.

  3. Skin: Cutaneous manifestations are common in GvHD and can serve as visible indicators of the disease's progression. Skin involvement often includes symptoms like rash, erythema, and blistering. At the tissue level, GvHD induces inflammation and damage to the skin's layers, disrupting the normal architecture.


According to The International Cone Marrow Transplant Registry (IBMTR), the severity of each areas are graded into few stages below:


Skin     

Stage 1: Maculopapular rash less than 25% of the body

Stage 2: Maculopapular rash 25% to 50% of the body

Stage 3: Generalized erythroderma

Stage 4: Generalized erythroderma with bullae


Liver

Stage 1: Bilirubin 2-3, AST 150-750

Stage 2: Bilirubin 3-6

Stage 3: Bilirubin 6-15

Stage 4: Bilirubin >15


GI system

Stage 1: Diarrhea >500 cc/day

Stage 2: Diarrhea >1000 cc/day

Stage 3: Diarrhea > 1500 cc/day

Stage 4: Diarrhea > 2000 cc/day or severe abdominal pain


International Cone Marrow Transplant Registry (IBMTR) Severity index:

  1. Mild: no liver or GI involvement, stage 1 skin involvement

  2. Moderate: stage 1-2 liver or GI involvement, stage 2 skin involvement

  3. Severe: stage 3 skin, liver, or GI involvement

  4. Life-threatening: stage 4 skin, liver, or GI involvement


Meanwhile, according to Glucksberg Grade, the severity of each areas are graded into few stages below:

  1. Mild: no liver or GI involvement, stage 1-2 skin involvement

  2. Moderate: stage 1 liver or GI involvement, stage 1-3 skin involvement

  3. Severe: stage 2-3 skin, liver, or GI involvement

  4. Life-threatening: stage 2-4 liver or GI involvement, stage 1-4 skin involvement


Understanding GVHD Management

Prophylactic treatment for GVHD is a critical component of the management strategy for patients undergoing HCT. These preventive measures are essential to reduce the risk of GVHD-related complications. Typically, treatment protocols vary between institutions, but a common approach involves a combination of cyclosporine and methotrexate administered for several months post-transplantation. Additionally, prophylaxis against bacterial, viral, and fungal infections is often implemented post-HCT to mitigate the heightened susceptibility to infections associated with the transplant process.


Treatment for established GVHD depends on the severity of symptoms and the organs involved. The primary goal is to suppress the immune response of donor T cells while striking a delicate balance to avoid compromising the beneficial graft-versus-tumor (GVT) response. Corticosteroids remain a cornerstone of GVHD treatment, with the choice and intensity of treatment modulated based on the grade and specific manifestations of GVHD. Grade 1 typically managed with topical steroids to control local symptoms. In cases of steroid-resistant disease, topical tacrolimus may be considered. Meanwhile, grade 2 or higher requires the addition of systemic steroids, commonly methylprednisolone at 2 mg/kg/day in divided doses. If gastrointestinal (GI) involvement is present, a nonabsorbable corticosteroid (such as budesonide or beclomethasone) may be added to systemic treatment. It's important to avoid steroids if there is evidence of a GI infection.


  • Tapering of Steroids: A gradual tapering of steroids over several months is crucial to prevent GVHD flares. Patients with chronic GVHD may need prolonged courses of steroids, often spanning 2 to 3 years, and some patients may require lifelong treatment. Octreotide, aimed at reducing diarrhea, can be added in certain cases.

  • Additional Agents: Beyond steroids, other agents may be added to the treatment regimen, including mycophenolate, etanercept, pentostatin, monoclonal antibodies, sirolimus, alpha-1-antitrypsin, mesenchymal stromal cells, and extracorporeal photopheresis. However, the efficacy of these agents is not entirely clear, and their use may vary based on individual patient factors.

  • Cyclosporine in Chronic GVHD: In cases of chronic GVHD, cyclosporine may be added to the treatment regimen to decrease the dosage and duration of steroid therapy.


Understanding GVHD Prevention

Preventing GVHD in the context of stem cell therapy requires a nuanced understanding of the immune dynamics involved in allogeneic stem cell transplantation. Here's a more detailed exploration of the key strategies:


1. Donor Selection: The selection of an appropriate donor is a critical first step. Compatibility in human leukocyte antigens (HLA) between the donor and recipient is vital. The closer the match, the lower the likelihood of the recipient's immune system recognizing the donor cells as foreign and triggering an immune response leading to GVHD. Advanced techniques, such as HLA typing, play a pivotal role in identifying suitable donors.

2. Pharmacological Prophylaxis: Immunosuppressive medications are central to preventing GVHD. Calcineurin inhibitors like cyclosporine or tacrolimus are often used to suppress the activity of T cells from the donor graft, preventing them from attacking the recipient's tissues. Methotrexate, an antimetabolite, is commonly combined with calcineurin inhibitors to further reduce the risk of GVHD.

3. Ex Vivo T-Cell Depletion: Ex vivo T-cell depletion involves selectively removing T cells from the donor graft before transplantation. This technique aims to reduce the number of potentially harmful T cells that could cause GVHD while preserving the stem cells responsible for the therapeutic effect. However, this approach comes with challenges, such as delayed immune reconstitution and increased susceptibility to infections due to a compromised immune system.

4. Conditioning Regimen: The conditioning regimen, comprising pre-transplant chemotherapy and/or radiation therapy, is carefully tailored to strike a delicate balance. Sufficient intensity is required to prepare the recipient's immune system for the incoming stem cells and prevent rejection, but excessive intensity can lead to an inflammatory response that triggers GVHD. Personalizing the conditioning regimen based on the patient's health and the nature of the underlying condition is essential.

5. Advanced Technologies: Emerging technologies bring exciting possibilities for GVHD prevention. Gene editing techniques allow for precise modification of donor cells to minimize their potential to cause GVHD. Regulatory T cell (Treg) infusions, which are immune cells with suppressive properties, offer another avenue to control the immune response and mitigate GVHD risk. These advanced approaches hold promise for more targeted and effective GVHD prevention.


In conclusion, GVHD prevention in stem cell therapy requires a comprehensive and individualized approach that integrates donor selection, pharmacological prophylaxis, advanced techniques, and personalized conditioning regimens. Achieving a delicate equilibrium between preventing rejection and minimizing GVHD risk is a dynamic challenge, and ongoing research aims to refine these strategies for enhanced safety and efficacy in the realm of stem cell transplantation.


Reference:

Current issues in chronic graft-versus-host disease (2014)

Severity Index for grading acute graft-versus-host disease: retrospective comparison with Glucksberg grade (1997)

Graft-versus-host disease (2009)

State-of-the-art acute and chronic GVHD treatment (2015)

Chronic graft-versus-host disease (2003)

Graft-Versus-Host Disease (2022)

 

Discover the Science behind Regenerative Medicine

in our upcoming Mini Fellowship happening globally:


IFAAS Mini Fellowship (Hands-On)

Anti-Aging Stem Cell, SVF Therapy & Regenerative Medicine




March 17, 2024 - Seoul, South Korea - [Register Now]


 

More Upcoming Global Events


bottom of page