Immunotherapy in Autoimmune Diseases:

Current Advances and Future Directions

 

Mukund M. Pache*, Rutuja R. Pangavhane

Department of Pharmacy, K.V.N. Naik S. P. Sanstha's, Institute of Pharmaceutical Education and Research, Nashik, 422002, Maharashtra, India.

*Corresponding Author E-mail: mukundpache918@mail.com

 

ABSTRACT:

Immunotherapy represents a novel approach for managing autoimmune diseases by targeting specific immune cells, cytokines, and checkpoints rather than broadly suppressing the immune system. Corticosteroids and disease-modifying antirheumatic drugs (DMARDs) remain commonly used treatments despite challenges such as lack of specificity and significant adverse effects. Immunotherapies, including monoclonal antibodies (mAbs), CAR-T cells, and immune checkpoint inhibitors (ICIs), address the underlying causes of immune system dysfunction, offering enhanced effectiveness for conditions such as rheumatoid arthritis, multiple sclerosis, systemic lupus erythematosus, and psoriasis. Emerging therapies include next-generation CAR-T and CAR-NK cells, gene editing technologies such as CRISPR, and personalized medicine approaches. These innovations allow for precise targeting of autoreactive cells and customization of treatments to individual immune profiles. Although the advances are promising, safety issues, high costs, and the variability in patient response are still the major hurdles. Key challenges include immune-related adverse events and infections, while economic barriers significantly limit accessibility. Additionally, variability in therapeutic outcomes highlights the need for biomarkers to predict responses and guide patient selection. AI-based tools could aid patient stratification and drug discovery, enhancing safety and efficacy in the future. Overall, sustained exploration in immunotherapy optimization and the introduction of new concepts are of utmost importance for the purpose of autoimmune disease management. Furthermore, these advancements hold the potential to extend remission duration and improve patient quality of life.

 

KEYWORDS: Immunotherapy, Autoimmune disease management, Monoclonal antibodies in autoimmunity, Cellular immunotherapy, Immune checkpoint modulation, Tolerance restoration strategies, Personalized immunotherapy approaches.

 

 


 

INTRODUCTION:

Autoimmune diseases encompass a range of conditions in which the immune system mistakenly targets the body's own tissues. It falsely attacks the self-antigen leading to chronic inflammation and tissue damage in different organs and systems.1 There are four main autoimmune diseases, namely, rheumatoid arthritis, multiple sclerosis, systemic lupus erythematosus, and type 1 diabetes, each of which is characterized by its own particular immune response that mistakenly treats self-antigens as the enemy2,3. Pathogenesis of autoimmune diseases is a very complicated process that includes the interactions of all three: genetic, environmental, and immunological factors. Genetic predispositions, such as polymorphisms in human leukocyte antigen (HLA) genes, are the most common ones that are known to increase the chances of developing autoimmunity, whereas the environmental ones, such as infections and toxins, can even onset the disease in the people that are genetically susceptible to it4,5. In the process of immune response, autoimmunity autoreactive T and B cells are the key players. These cells are involved in the production of autoantibodies and pro-inflammatory cytokines that cause specific damage in the tissues6. The malfunction in regulatory T cells (Tregs) and the improper cytokine production, such as interleukin (IL)-6, IL-17, and tumour necrosis factor (TNF), have been attributed to the cause and the advancement of several autoimmune diseases7,8. Regular methods for treating autoimmune conditions involve taking immunosuppressants, for example, corticosteroids, disease-modifying antirheumatic drugs (DMARDs), and non-steroidal anti-inflammatory drugs (NSAIDs). These medications are predominantly used to bring about inflammation control and ease symptoms. Thus, they have inherent drawbacks, which frequently manifest in the form of side effects and insufficient directing of disease mechanisms. This may cause generalized immunosuppression which generally makes one vulnerable to infections and malignancies9. Lately, immunotherapy has come to light as a potential solution against the traditional means, which can be more precise in autoimmune diseases. Immunotherapies, namely monoclonal antibodies, immune checkpoint inhibitors, and cell-based therapies, are exclusively aimed at the immune pathways that are involved in the development of the disease, thereby, minimizing the risk of an overall immunosuppression and increasing therapeutic efficacy10,11. Discoveries in the way diseases of the immune system work have led to therapies such as TNF inhibitors, IL-6 blockers, and T-cell-targeted medicines being used and these have been noted for significant improvements in autoimmune patients12,13. Moreover, recent methods such as CAR-T cell therapy and gene editing technologies being studied are having the effect of providing personalized and long-lasting treatment for patients suffering from autoimmune conditions14. In summary, immunotherapy marks a revolutionary change to the management of autoimmune diseases, addressing the main weaknesses of standard treatments and bringing the hope for targeted, effective, and durable therapies to the horizon.

 

1.     Overview of Immunotherapy Approaches in Autoimmune Diseases15,16:

Today, rather than using general immunosuppression, immunotherapy strategies in autoimmune diseases developed towards the targeted immunomodulation of particular pathways. These therapies attack solely immune cells, interleukins, and signal pathways affected in autoimmunity when they dysregulated the immune system. The following is a summary of the main methods of immunotherapy implemented or investigated for autoimmune diseases.

 

 

Figure 1. Overview of Immunotherapy Approaches in Autoimmune Diseases

 

1.1 Monoclonal Antibodies (mAbs):

Monoclonal antibodies (mAbs) are a fundamental part of immunotherapy for autoimmune diseases, and they are also, in addition to the above, used in cancer therapy and organ transplantation. Autoimmune diseases encompass a range of conditions in which the immune system mistakenly targets the body's own tissues. TNF inhibitors, such as infliximab and adalimumab, are the first mAbs that were introduced for the treatment of disorders, which are commonly known as RA or Crohn's disease, and prove to be effective in these disorders through their effect in neutralizing TNF, a pro-inflammatory cytokine. Other mAbs target interleukin-6 (IL-6), another cytokine involved in the autoimmune process17,18. Tocilizumab, an IL-6 receptor antagonist, has proven effective in relieving rheumatoid arthritis symptoms in patients unresponsive to TNF inhibitors19. Rituximab, a CD20-directed mAb, causes B cells with auto antigen specificity to be depleted, thus targeting pathogenic auto-reactive B cell populations in multiple sclerosis (MS) and systemic lupus erythematosus (SLE) respectively20–22. As a consequence of this blockade, mAbs may specifically cure or reverse the processes in the disease without the massive immune system suppression usually exhibited by conventional therapies.

 

1.2 Immune Checkpoint Inhibitors (ICIs):

Immune checkpoint inhibitors (ICIs), originally created for treating cancer, have come to be in investigations for autoimmune disease because of their concept to modulate immune responses. ICIs, such as anti-CTLA-4 and anti-PD-1 antibodies, inhibit T-cell signaling pathways, intensifying immune responses in cancer but requiring fine-tuning for autoimmune applications23. Meanwhile, instead of stimulating the immune response as in cancer, their application in autoimmunity emphasizes rebalancing the active immune responses. Initial studies imply that ICIs might facilitate a reduction in autoimmune disorders by either ameliorating the regulatory T-cell (Treg) function or by inducing exhaustion in autoreactive T cells24–26. Nevertheless, their application in autoimmunity is qualified by fine-tuning to limit immune activation, which is a frequent adverse effect ICIs recipients suffering from cancer experience.

 

1.3 CAR-T Cell Therapy:

The Chimeric antigen receptor T-cell (CAR-T) therapy originated as cancer therapy spurred by its potential to attack cancer cells. The technique has lately been applied in the treatment of autoimmune disorders as well. This method of treatment is called active cell therapy, and it entails modifying the T cells of a patient such that they bring out receptors that precisely recognize and destroy the immune cells. The preliminary studies have indicated that CAR-T cells selective for autoreactive B cells are capable of decreasing the condition's activity in situations like SLE and myasthenia gravis27,28. Early clinical studies to assess the effectiveness of CAR-T therapy in the remission of severe forms of autoimmune diseases are in progress29. This highly individualized way of dealing has been a success in cases that are not responsive to regular treatment and opens new possibilities for autoimmune therapy30,31.

 

1.4 T-cell and B-cell Modulation:

T-cell and B-cell modulation therapies are interventions that aim to restore the balance of the immune system by the regulation of these key cells in the body. Abatacept, a CTLA-4-Ig fusion protein, is the favourite tool of T-cell modulation as it locks the T-cell costimulatory signals necessary for T-cell activation and ameliorates the severity of RA and SLE32. This leads to a mechanism of inhibition of the initial immune responses that are normally associated with tissue damage. B-cell modulation therapy, involving medications like belimumab that are directed against B-cell survival signalling molecules, results mainly in the mitigation of B-cell survival and activity in SLE33. These therapies offer personalized therapy for immune modulation and thus reduce off-target effects to the only degree necessary for attenuation of autoreactive T and B cells34.

 

1.5 Other Immunotherapies:

Besides mAbs, ICIs, and cellular therapies, other types of immunotherapies are being investigated to treat autoimmune diseases. Hematopoietic stem cell transplantation (HSCT) has been applied to the impact on serious autoimmune cases with the intention of resetting the immune system. Thus, the option is being further valued in MS and systemic sclerosis, as per the review35. Some of the gene therapy techniques being developed aim to fix the inherited genetic defects that cause autism or reprogram immune cells to tolerate self-antigens; these approaches are at present still in the developing stage36. Reflex-based peptide therapies, which are developed to cause tolerance to particular autoantigens, have also ensured positive results. In particular, MS patients have shown high efficacy of the therapy. These developing strategies represent a great range of potentialities that immunotherapy has to redesign the whole autoimmunity treatment in new ways37. The range of different immunotherapy approaches for autoimmune diseases creates the hope that it may be possible to develop more targeted, effective, and durable treatment. Even though every technique possesses certain distinct complications, including side effects and response variability, research and clinical trials that are in progress are aiding these therapies to be exact and the best for treating autoimmune conditions38.

 

2.     Mechanisms of Action and Immunological Targets:

In autoimmune diseases, immunotherapy works by the body to cut the immune cells, cytokines, and immune checkpoints which are the main factors in the pathology of the disease. A course of treatment is then designed to target a certain area or areas responsible for the immune deficiency involved in autoimmunity and, consequently, the issues of the disease. This also comes with the added benefit of fewer defects.

 

 

Figure 2.  Overview of Immunotherapy Approaches in Autoimmune Diseases

 

2.1 Targeting B Cells and T Cells:

B cells are one of the main targets of immunotherapy in autoimmune diseases, as they are responsible for the production of self-attacking antibodies and the presentation of antigens to T cells. Treatments like rituximab, a monoclonal antibody that aims at the CD20 on the B cells, depletes the B cells, therefore, reducing autoantibody production and consequently breaking the inflammation cycle. This strategy has demonstrated effectiveness in cases such as multiple sclerosis (MS) and systemic lupus erythematosus (SLE)20,21,39. Not only does B-cell depletion take away the source of autoantibodies, but it also prevents the activation of autoreactive T cells, which is an additional merit in situations of autoimmunity40. As for T cells, therapies are developed to interfere with their activation and function, mainly because these cells are the main drivers of diseases like rheumatoid arthritis (RA) and psoriasis. Abatacept, a fused protein of CTLA-4-Ig, blocks the signal that is needed for T-cell activation. Therefore, it curbs the immune response32. This specific suppression of T-cell activation can reduce inflammation without causing the whole body to become immunosuppressed. Likewise, CAR-T cell therapy has been applied in autoimmunity, where T cells are altered to recognize and destroy autoreactive immune cells, thus giving a very personalized method to regulate immune responses27.

 

2.2 Cytokine Modulation: IL-6, IL-17, and TNF:

Cytokines, which are the main makers of autoimmune diseases, exhibit their roles as signalling molecules that cause inflammation and call-in immune cells to places where there is tissue damage. IL-6 is a pro-inflammatory cytokine, and monoclonal antibodies such as tocilizumab are directed at it. Therefore, the efficacy of this drug in RA has been demonstrated through the reduction of IL-6 levels, and inflammation is restricted19. IL-17 is another cytokine that has been linked to autoimmune diseases such as ankylosing spondylitis and psoriasis and is the target of therapies like secukinumab. IL-17 blockade leads to the reduction of inflammatory cell recruitment to tissues and thus, the inflammatory cascade is mainly addressed41,42. Furthermore, TNF inhibitors like infliximab and adalimumab sink their teeth into TNF, which is the main cytokine in autoimmune diseases like Crohn’s disease and RA, hence, the inflammation is managed at the source42.

 

2.3 Immune Checkpoint Inhibition and CAR-T Engineering

Immune checkpoint inhibitors (ICIs) are utilized to control the hyper-immune reactions observed in autoimmunity. They do this by modulating immune checkpoints like CTLA-4 and PD-1, which usually operate as the "brakes" of the immune system43. In contrast to cancer, autoimmune diseases have a different therapeutic aim where the T cells are activated through checkpoint inhibition44. Instead, in autoimmunity, the therapy might be directed to up-regulate the checkpoint pathways to induce tolerance or exhaustion in autoreactive T cells, which restores immune homeostasis25,45.

 

CAR-T cell therapy is an innovative strategy that incorporates the concepts of checkpoint blockade and cell engineering in autoimmunity. In this technique, T cells are genetically engineered to produce chimeric antigen receptors (CARs) that are capable of identifying specifically the antigens on autoreactive cells. As a result, CAR-T cells can be involved in the clearance of pathogenic immune cells selectively and this will lead to reduced disease activity46. By the introduction of the CAR-T cells that will carry the inhibitory molecules, the researchers are trying to develop the CAR-T cells that will not only serve the function of the targeted immune modulators but also perform the tolerance agent within the immune system47,48.

 

These therapies represent the movement from traditional medicine towards precision medicine in autoimmune diseases by focusing on particular immune components and pathways that are key to the development of the disease. These therapies are the ones that solve the roots of the disease more precisely, which is why they have the potential to persist.

 

3.     Case Studies and Clinical Trials in Specific Autoimmune Diseases:

Immunotherapy has greatly progressed in the scope of autoimmune disease management with the introduction of specific treatments that can outperform the previous ones. Below is a review of the most notable immunotherapy strategies applied in the treatment of rheumatoid arthritis, multiple sclerosis, systemic lupus erythematosus, and psoriasis.

 

3.1 Rheumatoid Arthritis:

Rheumatoid arthritis (RA) is a chronic autoimmune disease that affects the joints and the surrounding tissues. Monoclonal antibody drugs and immune checkpoint drugs have totally changed the treatment of RA. The CD20 antibody rituximab was initially approved in RA patients unresponsive to TNF inhibitors. Through the depletion of B cells, rituximab has the ability to control disease progression and among the clinical trials undertaken, it has been proven that it maintains these effects49,50. In a major study, patients who were treated with rituximab had the advantage of decreases in the degree of the illness along with improvements in their life quality compared to the placebo group51. Furthermore, one of the most encouraging treatments for RA is the use of ICIs to modulate the immune response. Inflammation inhibitor medications such as ICIs, which are mainly used in cancer treatment, can be applied in bulimia to show positive changes in the course of immune dysregulation. However, the concern regarding the accentuation of autoimmune disease complications dictates the use of ICIs in RA only with very close monitoring23,52. Currently, clinical studies analyzing the optimal immune checkpoint optical in autoimmune disorders, are still in the process.

 

3.2 Multiple Sclerosis :

Multiple sclerosis (MS) is an autoimmune disease where the destruction (by the immune cells) of the central nervous system leads to demyelination and neurodegeneration. Medications such as ocrelizumab and alemtuzumab have been quite effective in preventing the onset of disease through the targeting of B and T cells. Ocrelizumab, which is a monoclonal antibody directed at B-cells, has been shown to be effective in both relapsing-remitting MS and primary progressive MS by reducing the number of relapses and slowing down the progression of disability53. The OPERA and ORATORIO clinical trials confirmed the satisfactory results of ocrelizumab as it led to lower relapse rates and fewer MRI-detected lesions as compared to interferon beta-1a54,55.

 

Alemtuzumab, reaching to CD52 present on T and B cells, triggers long-term lymphocyte deletion that can end up in durable remission in the relapsing-remitting MS.  The CARE-MS studies have demonstrated that alemtuzumab is very successful in cutting down relapse rates and increasing disability outcomes in comparison to traditional disease-modifying therapies56. On the other hand, alemtuzumab is also connected with a high risk of secondary autoimmune disorders, including thyroid disease. Hence, extreme caution should be applied to the selection and supervision of patients57.

 

3.3 Systemic Lupus Erythematosus (SLE):

Systemic lupus erythematosus (SLE) is a chronic immune-mediated non-organ specific disorder. Belimumab, a monoclonal antibody (mAb) that blocks B-cell activating factor (BAFF), is only one of the approved biologics solely for SLE. BLISS clinical trials, along with others, have proven that belimumab reduces disease activity and flare rates in patients suffering from active SLE and, therefore, can be utilized as an alternative to those who fail to show improvement in the conventional treatments33. Apart from that, Belimumab has been seen to improve the condition of patients suffering from lupus nephritis, the most severe complication of SLE involving kidney failure58.

 

Studies on novel drugs for treating lupus nephritis are currently conducted, among them are mAbs targeting the IL-12 and IL-23 pathways under investigation. A study conducted at the National University Hospital of Singapore showed that the down-regulation of IL-23 among SLE subjects served to reduce renal inflammation, hence giving a green light to therapeutic scientists in terms of research and development of a treatment for this condition59. Another promising investigational technique for it is the modified version of CAR T-cell therapy that was tested in preclinical trials and showed efficient and prolonged disease remission27.

 

3.4 Psoriasis and Psoriatic Arthritis

Psoriasis is a long-lasting inflammation of the skin that frequently cooccurs with psoriatic arthritis, which affects the joints. IL-17 and IL-23 cytokines are the main reasons for psoriasis progression and the treatments that are directed against these pathways are very successful. Secukinumab, one of the most widely investigated IL-17A inhibitors, is the best-studied drug for psoriasis and psoriatic arthritis. In clinical trials, secukinumab demonstrated a significant improvement in skin lesions and joint symptoms, and the majority of patients achieved complete or near-complete skin clearance60. In the ERASURE and FIXTURE studies, secukinumab brought about marked decreases in psoriasis severity scores compared to placebo and standard therapies61. IL-23 inhibitors (e.g., guselkumab) are also effective in psoriasis and psoriatic arthritis. Guselkumab, by efficiently targeting the IL-23/Th17 axis, has proven very impressive in preventing the symptoms and the development of joint damage62. The studies emphasize the durability of the response with IL-23 inhibition, as patients kept attaining remission after the long follow-up63,64.

 

Utilization of target immunotherapy in immune-mediated disorders has shown considerable efficacy in a wide variety of autoimmune diseases. Among others, B-cell-depleting therapies have been used in MS, and cytokine inhibitors are given to psoriasis patients; hence, these innovations pave the way for patients who have been unamenable to treatments earlier. Yet, there are problems concerning the side effects of therapies like alemtuzumab and in order to find the right patient for a treatment and improve the long-term outcomes, it is necessary to do more research.

 

4.     Current Challenges in Immunotherapy for Autoimmune Diseases:

Even though there have been incredible developments in immunotherapy for autoimmune diseases, these methods cannot be used widely because of some limitations. Safety issues, the expenses of the treatments, and the mixed success in different patients are all a part of the picture here, and therefore clinical application of these therapies might be tricky.

 

 

Figure 3.  Current Challenges in Immunotherapy for Autoimmune Diseases

 

4.1 Safety Concerns:

Adverse effects, in general, specifically immune-related adverse events (irAEs), are one of the most challenging parts of immunotherapy. Better outcomes with immunotherapies like ICIs and T-cell-targeted therapies may be offset by immune system overactivity or inactivity that results in infections, malignancies, and autoimmune symptoms65. For example, chemotherapy like alemtuzumab, a medicine used for treating multiple sclerosis (MS), has been found to result in secondary autoimmune conditions which are like thyroiditis and nephropathy57. In parallel, a similar case of B-cell depletion, for example, of the use of rituximab and ocrelizumab, which highly increases the risk of infection for patients with already weak immune systems53. Thus, these medications are not suitable for all patients and need regular visits by a doctor. Indeed, they might also restrict the population which can use immunotherapy drugs.

 

4.2 Cost and Accessibility:

The high expenses of immunotherapy are a severe obstacle, thereby affecting patients and health care systems. Immunotherapy, which includes drugs like monoclonal antibodies, CAR-T cell therapies, and ICIs, costs a lot and patients might incur huge treatment bills over time66. An economic evaluation of rheumatoid arthritis indicated that the TNF inhibitors and IL-6 inhibitors, which are biologics, have proven to be very strenuous due to the demands they place on the budgets of health systems12,63. However, tipping the scales toward the unemployed regions, it is the affordable settings that are less likely due to the lack of treatment of k-RAS. Consequently, only patients in the low-budget zones get to take advantage of the immunotherapies67.

 

4.3 Patient Variability and Biomarkers :

The immunotherapy reactions to different patients are very diverse and the outcomes still cannot be predicted. Elements like genetic predisposition, the severity of the disease, and the immune profile determine the patient's response to the treatment thus resulting in variable efficacy and side effects68. For instance, a section of patients suffering from systemic lupus erythematosus (SLE) does not respond to belimumab, which means that some groups of patients may be more appropriate candidates33,69. Identifying reproducible biomarkers for treatment response prediction and patient stratification is key but still at work. The success in biomarker identification by means of cytokine profiling or genetic markers is encouraging but needs further confirmation in order to be applied to medical practice70.

 

Immunotherapy, although carrying the potential of a breakthrough in autoimmune ailments, still faces many challenges such as safety risks, treatment costs, and patients' varied responses. Overcoming these challenges by means of specific research and the implementation of better patient stratification will be a prerequisite for the complete use of immunotherapy for autoimmune conditions71.

 

5.     Future Directions and Emerging Therapies:

The area of immunotherapy for autoimmune diseases is an ever-evolving sector with novel approaches that exhibit the ability to optimize precision and effectiveness. Breaking through cellular therapies, personalized medicine, gene editing, and Al (or artificial intelligence) opens the way for the potential solution to the constraints that still haunt current therapies and bring good future paths.

 

Figure 4.  Future Directions and Emerging Therapies

 

5.1 Next-Generation CAR-T and CAR-NK Cells:

Chimeric antigen receptor T-cell (CAR-T) therapy, which has its roots in cancer treatment, is being further evolved for autoimmune diseases, while next-generation CAR-T and CAR-NK (natural killer) cells are under development to improve accuracy in the identification of autoreactive immune cells. In contrast, traditional CAR-T cells are engineered to target specific antigens expressed on tumour cells, and the next generation of CAR-T cells are built to specifically attack autoreactive B or T cells, which are responsible for autoimmune pathology27. These CAR-T cells have the ability to cause the disease to completely go into remission in the case of systemic lupus erythematosus (SLE) and multiple sclerosis (MS). Besides CAR-NK cells which are designed to target the tumor cells, NK cells modified to recognize the autoreactive cells are a new technology. They exhibit a safer profile owing to a reduced likelihood of cytokine release syndrome72. Preclinical studies and early trials confirmed that CAR-NK cells could be safer while still retaining sufficient efficacy in the autoimmune context73.

 

5.2 Personalized Medicine Approaches:

Personalized medicine has become more useful in immunotherapy, using genetic and molecular profiling to adapt the treatment to the particular patient. Genetic profiling, which includes HLA genotyping, is a method that detects patients who are more likely to experience positive responses to particular therapies whereas molecular biomarkers provide a means to evaluate disease activity as well as to predict the treatment response74. The development of single-cell sequencing and multi-omics methods has made it possible for in-depth immune cell analysis, thus, fostering the development of individual therapies. Clinicians can now choose the most effective treatment based on the molecular characteristics of patients with different immune signatures, who might respond differently to therapies like rituximab or IL-17 inhibitors9,75. Linking medications to individual immune profiles through personalized immunotherapy means better results and fewer side effects.

 

5.3 Combination Therapies:

Combination therapy strategies are more and more investigated as a way to enhance immunotherapy efficacy by targeting multilateral immune pathways. The combination of biologics (TNF blockers, IL-6, or IL-17 inhibitors) has proven it can overcome refractory cases of rheumatoid arthritis and psoriatic arthritis successfully in some of the patients. A different way to combine it with immunotherapy is to add regular drugs like corticosteroids or DMARDs to get a synergistic effect, so the amount of each medicine will be less and the side effects will be reduced. Clinical trials are already being conducted to test the safety and effectiveness of these combinations, with the initial data showing that if wisely selected, combo sites can have a bigger effect76,77.

 

5.4 Gene Editing (e.g., CRISPR) :

Gene editing technologies, particularly CRISPR, have provided new opportunities to tackle autoimmunity at the genetic level. The introduction of CRISPR technology makes it possible to change the immune cell genes so that the mutations that are the causes of autoimmune diseases can be corrected or the cells can be reprogrammed for tolerance. To illustrate, scientists are looking into the CRISPR-mediated inactivation of genes which are the cause of autoreactive T or B cell receptors, and thus, the cells responsible for autoimmune attacks can be reduced78. Furthermore, CRISPR can also be used to augment regulatory T cells (Tregs) by editing genes that induce their suppressive functions, and as a result, the immune system is brought back to balance79. Nevertheless, these gene-editing techniques, which are still in experimental stages, may eventually become permanent solutions to autoimmune diseases by targeting the underlying genetic mutation.

 

5.5 Artificial Intelligence and Machine Learning:

AI and machine learning come with vast possibilities for the use of immunotherapy as they allow scientists to discover the drugs for it and predict treatment outcomes and new targets in the immune system one of which is by means of the technology of AI. Machine learning applications to medicine can be trained on immense datasets to uncover the patterns that are associated with successful or unsuccessful therapies, thus making predictions that can be used in patient stratification and risk assessment80. In the case of, for instance, machine learning systems can blend clinical, genetic, and immunological data to "identify the patients who are likely to respond to" the therapy of T cells or checkpoint inhibitors. Besides that, Artificial Intelligence is currently employed to unveil new targets for drugs by inspecting molecular pathways involving autoimmunity which will lead to the distillation of cutting-edge immunotherapies81,82. The improvements brought by AI to immunotherapy will most likely cause its development to a stage where it really is a tool doctors can make use of for patient-tailored therapy.

 

CONCLUSION:

Immunotherapy has turned into a new paradigm in the treatment of autoimmune diseases by supplying options that are highly specific and disease-modified and can tackle the immune imbalance along with the main problem rather than just symptom relief. Therapies like monoclonal antibodies (mAbs), CAR-T cells, and immune checkpoint inhibitors (ICIs) have manifested effectiveness in autoimmune diseases such as rheumatoid arthritis, multiple sclerosis, systemic lupus erythematosus, and psoriasis by switching off the key molecules of the immune system. New therapies, including CAR-NK cells, gene editing, and personalized medicine, provide even better stereotyping of autoreactive cells and the ability of the immune system profile to adapt. Thus, a comprehensive cure takes place. Nevertheless, serious obstacles have been encountered, such as the risk of immune-related dreadful side effects, a heavy cost burden, and baseline differences in patient outcomes. To go beyond these drawbacks the focus should be on ongoing research to make safety profiles optimal, costs reduced, and if possible, markers should be used to choose the most appropriate therapy for each patient.

 

Immunotherapy is a new development in the management of autoimmune diseases that shows a lot of potential. It has the possibility of sustainable remission for patients considerably higher and their lives will be improved due to the remarkable technological and scientific progress that has been made. By continuous investments in innovation and research, we will be able to reap these benefits. Hence, immunotherapy will be the most effective tool for autoimmunity disease management in the future.

 

CONFLICT OF INTEREST:

The authors declare no conflict of interest.

 

REFERENCE:

1.   Rose NR, Bona C. Defining criteria for autoimmune diseases (Witebsky’s postulates revisited). Immunology Today. 1993;14(9):426–430; doi: 10.1016/0167-5699(93)90244-F.

2.    Marrack P, Kappler J, Kotzin BL. Autoimmune disease: why and where it occurs. Nat Med. 2001;7(8):899–905; doi: 10.1038/90935.

3.    Davidson A, Diamond B. Autoimmune Diseases. Mackay IR, Rosen FS. eds. N Engl J Med. 2001;345(5):340–350; doi: 10.1056/NEJM200108023450506.

4.    Huseby ES, Liggitt D, Brabb T, et al. A Pathogenic Role for Myelin-Specific Cd8+ T Cells in a Model for Multiple Sclerosis. The Journal of Experimental Medicine. 2001;194(5):669–676; doi: 10.1084/jem.194.5.669.

5.    Mohan C, Putterman C. Genetics and pathogenesis of systemic lupus erythematosus and lupus nephritis. Nat Rev Nephrol. 2015;11(6):329–341; doi: 10.1038/nrneph.2015.33.

6.    Alunno A, Carubbi F, Giacomelli R, et al. Cytokines in the pathogenesis of rheumatoid arthritis: new players and therapeutic targets. BMC Rheumatol. 2017;1(1):3; doi: 10.1186/s41927-017-0001-8.

7.    Weaver CT, Hatton RD, Mangan PR, et al. IL-17 Family Cytokines and the Expanding Diversity of Effector T Cell Lineages. Annu Rev Immunol. 2007; 25(1): 821–852; doi: 10.1146/annurev.immunol.25.022106.141557.

8.    Tanaka T, Narazaki M, Kishimoto T. IL-6 in Inflammation, Immunity, and Disease. Cold Spring Harbor Perspectives in Biology. 2014; 6(10): a016295–a016295; doi: 10.1101/cshperspect.a016295.

9.    Smolen JS, Aletaha D, McInnes IB. Rheumatoid arthritis. The Lancet. 2016; 388(10055):2023–2038; doi: 10.1016/S0140-6736(16)30173-8.

10.  Bluestone JA, Herold K, Eisenbarth G. Genetics, pathogenesis and clinical interventions in type 1 diabetes. Nature. 2010; 464(7293): 1293–1300; doi: 10.1038/nature08933.

11.  Bluestone JA, Herold K, Eisenbarth G. Genetics, pathogenesis and clinical interventions in type 1 diabetes. Nature. 2010; 464(7293): 1293–1300.

12.  Smolen JS, Landewé R, Bijlsma J, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2016 update. Ann Rheum Dis. 2017; 76(6): 960–977; doi: 10.1136/annrheumdis-2016-210715.

13.  Burmester GR, Pope JE. Novel treatment strategies in rheumatoid arthritis. The Lancet. 2017; 389(10086): 2338–2348; doi: 10.1016/S0140-6736(17)31491-5.

14.  June CH, O’Connor RS, Kawalekar OU, et al. CAR T cell immunotherapy for human cancer. Science. 2018;.359(6382): 1361–1365; doi: 10.1126/science.aar6711.

15.  Kumar A, Singh K, Kumar K, et al. Immunotherapy in Cancer Treatment: Harnessing the Power of the Immune System. RJPDFT 2024;107–112; doi: 10.52711/0975-4377.2024.00017.

16.  Mathayan M, Suresh A, Balamurugan R, et al. Immune Stimulation effects of Pongamia pinnata extracts, an In vitro Analysis. Rese Jour of Pharm and Technol. 2020; 13(1): 308; doi: 10.5958/0974-360X.2020.00062.1.

17.  Elliott MJ, Maini RN, Feldmann M, et al. Randomised double-blind comparison of chimeric monoclonal antibody to tumour necrosis factor α (cA2) versus placebo in rheumatoid arthritis. The Lancet. 1994; 344(8930): 1105–1110; doi: 10.1016/S0140-6736(94)90628-9.

18.  Sandborn WJ, Hanauer SB, Katz S, et al. Etanercept for active Crohn’s disease: a randomized, double-blind, placebo-controlled trial. Gastroenterology. 2001; 121(5): 1088–1094.

19.  Gabay C, Emery P, Van Vollenhoven R, et al. Tocilizumab monotherapy versus adalimumab monotherapy for treatment of rheumatoid arthritis (ADACTA): a randomised, double-blind, controlled phase 4 trial. The Lancet. 2013; 381(9877): 1541–1550; doi: 10.1016/S0140-6736(13)60250-0.

20.  Hauser SL, Waubant E, Arnold DL, et al. B-Cell Depletion with Rituximab in Relapsing–Remitting Multiple Sclerosis. N Engl J Med. 2008; 358(7): 676–688; doi: 10.1056/NEJMoa0706383.

21.  Merrill JT, Neuwelt CM, Wallace DJ, et al. Efficacy and safety of rituximab in moderately‐to‐severely active systemic lupus erythematosus: The randomized, double‐blind, phase ii/iii systemic lupus erythematosus evaluation of rituximab trial. Arthritis & Rheumatism. 2010; 62(1): 222–233; doi: 10.1002/art.27233.

22.  Wahono CS, Poeranto S. Gingivitis on Systemic Lupus Erythematosus (SLE) patients: a pilot study. Age (yea rs). 2020;29:9–62.

23.  Fife BT, Bluestone JA. Control of peripheral T‐cell tolerance and autoimmunity via the CTLA‐4 and PD‐1 pathways. Immunological Reviews. 2008; 224(1): 166–182; doi: 10.1111/j.1600-065X.2008.00662.x.

24.  Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018; 359(6382): 1350–1355; doi: 10.1126/science.aar4060.

25.  Sharpe AH, Pauken KE. The diverse functions of the PD1 inhibitory pathway. Nat Rev Immunol. 2018; 18(3):.153–167; doi: 10.1038/nri.2017.108.

26.  Mandlik YR. Immunotherapy, a type of cancer treatment. Research Journal of Pharmaceutical Dosage Forms and Technology. 2020; 12(4): 282–284; doi: 10.5958/0975-4377.2020.00046.4.

27.  Ellebrecht CT, Bhoj VG, Nace A, et al. Reengineering chimeric antigen receptor T cells for targeted therapy of autoimmune disease. Science. 2016; 353(6295): 179–184; doi: 10.1126/science.aaf6756.

28.  Abdalhadi HM, Chatham WW, Alduraibi FK. CAR-T-Cell Therapy for Systemic Lupus Erythematosus: A Comprehensive Overview. IJMS. 2024; 25(19): 10511; doi: 10.3390/ijms251910511.

29.  Hartmann J, Schüßler‐Lenz M, Bondanza A, et al. Clinical development of CAR T cells—challenges and opportunities in translating innovative treatment concepts. EMBO Mol Med. 2017; 9(9): 1183–1197; doi: 10.15252/emmm.201607485.

30.  Bajirao Suryawanshi S, D Khaire R. Review on Car-T Cell Therapy for Cancer Treatment. IJT. 2023; 68–72; doi: 10.52711/2231-3915.2023.00008.

31.  Sen A, Kumar K, Khan S, et al. Current Therapy in Cancer: Advances, Challenges, and Future Directions. AJNER. 2024; 77–84; doi: 10.52711/2349-2996.2024.00016.

32.  Kremer JM, Westhovens R, Leon M, et al. Treatment of Rheumatoid Arthritis by Selective Inhibition of T-Cell Activation with Fusion Protein CTLA4Ig. N Engl J Med. 2003; 349(20): 1907–1915; doi: 10.1056/NEJMoa035075.

33.  Navarra SV, Guzmán RM, Gallacher AE, et al. Efficacy and safety of belimumab in patients with active systemic lupus erythematosus: a randomised, placebo-controlled, phase 3 trial. The Lancet. 2011; 377(9767): 721–731; doi: 10.1016/S0140-6736(10)61354-2.

34.  Amin AM, Muhammad Ansori AN, Kharisma VD, et al. T-Cell Epitope Vaccine Prediction Analysis Targeting Phosphoprotein (P) Rabies Virus Based on the Presence of HLA-I Alleles A, B, and C Loci Throughout Southeast Asia: An Immunoinformatics Study. RJPT. 2024; 2001–2008; doi: 10.52711/0974-360X.2024.00317.

35.  Burman J, Tolf A, Hägglund H, et al. Autologous haematopoietic stem cell transplantation for neurological diseases. Journal of Neurology. Neurosurgery and Psychiatry 2018;89(2):147–155.

36.  Zhong M-C, de Rosbo NK, Ben-Nun A. Multiantigen/multiepitope–directed immune-specific suppression of “complex autoimmune encephalomyelitis” by a novel protein product of a synthetic gene. The Journal of Clinical Investigation. 2002; 110(1): 81–90.

37.  Zubizarreta I, Flórez-Grau G, Vila G, et al. Immune tolerance in multiple sclerosis and neuromyelitis optica with peptide-loaded tolerogenic dendritic cells in a phase 1b trial. Proc Natl Acad Sci USA. 2019; 116(17): 8463–8470; doi: 10.1073/pnas.1820039116.

38.  Krienke C, Kolb L, Diken E, et al. A noninflammatory mRNA vaccine for treatment of experimental autoimmune encephalomyelitis. Science. 2021; 371(6525): 145–153; doi: 10.1126/science.aay3638.

39.  Adnan A, Perwitasari DA, Maliza R. Trend In the Publication of Study on Systemic Lupus Erythematosus (SLE) In 2012-2020 Period: A Bibliometric Analysis. RJPT. 2024; 1076–1082; doi: 10.52711/0974-360X.2024.00168.

40.  Cambridge G, Leandro MJ, Teodorescu M, et al. B cell depletion therapy in systemic lupus erythematosus: Effect on autoantibody and antimicrobial antibody profiles. Arthritis and Rheumatism. 2006; 54(11): 3612–3622; doi: 10.1002/art.22211.

41.  Baeten D, Sieper J, Braun J, et al. Secukinumab, an Interleukin-17A Inhibitor, in Ankylosing Spondylitis. N Engl J Med. 2015; 373(26): 2534–2548; doi: 10.1056/NEJMoa1505066.

42.  Langley RG, Elewski BE, Lebwohl M, et al. Secukinumab in Plaque Psoriasis — Results of Two Phase 3 Trials. N Engl J Med. 2014; 371(4): 326–338; doi: 10.1056/NEJMoa1314258.

43.  Kittai AS, Oldham H, Cetnar J, et al. Immune checkpoint inhibitors in organ transplant patients. Journal of Immunotherapy. 2017; 40(7): 277–281.

44.  Baksh K, Weber J. Immune Checkpoint Protein Inhibition for Cancer: Preclinical Justification for CTLA-4 and PD-1 Blockade and New Combinations. In: Seminars in Oncology Elsevier; 2015; pp. 363–377.

45.  Rowshanravan B, Halliday N, Sansom DM. CTLA-4: a moving target in immunotherapy. Blood, The Journal of the American Society of Hematology. 2018; 131(1): 58–67.

46.  Chen Y, Sun J, Liu H, et al. Immunotherapy Deriving from CAR-T Cell Treatment in Autoimmune Diseases. Journal of Immunology Research. 2019; 2019: 1–9; doi: 10.1155/2019/5727516.

47.  Kebriaei P, Kelly SS, Manuri P, et al. CARs: driving T-cell specificity to enhance anti-tumor immunity. Frontiers in bioscience (Scholar edition). 2012; 4: 520.

48.  Zhang Q, Lu W, Liang C-L, et al. Chimeric antigen receptor (CAR) Treg: a promising approach to inducing immunological tolerance. Frontiers in Immunology. 2018; 9: 2359.

49.  Edwards JCW, Szczepański L, Szechiński J, et al. Efficacy of B-Cell–Targeted Therapy with Rituximab in Patients with Rheumatoid Arthritis. N Engl J Med. 2004; 350(25): 2572–2581; doi: 10.1056/NEJMoa032534.

50.  Emery P, Fleischmann R, Filipowicz‐Sosnowska A, et al. The efficacy and safety of rituximab in patients with active rheumatoid arthritis despite methotrexate treatment: Results of a phase IIB randomized, double‐blind, placebo‐controlled, dose‐ranging trial. Arthritis & Rheumatism. 2006; 54(5): 1390–1400; doi: 10.1002/art.21778.

51.  Cohen SB, Emery P, Greenwald MW, et al. Rituximab for rheumatoid arthritis refractory to anti–tumor necrosis factor therapy: Results of a multicenter, randomized, double‐blind, placebo‐controlled, phase III trial evaluating primary efficacy and safety at twenty‐four weeks. Arthritis and Rheumatism. 2006; 54(9): 2793–2806; doi: 10.1002/art.22025.

52.  Srivastava S, Patel S, Daharwal SJ, et al. Rheumatoid Arthritis: An Autoimmune Disease Prevalent in Females. Rese Jour of Pharm and Technol. 2016; 9(2): 170; doi: 10.5958/0974-360X.2016.00030.5.

53.  Hauser SL, Bar-Or A, Comi G, et al. Ocrelizumab versus Interferon Beta-1a in Relapsing Multiple Sclerosis. N Engl J Med. 2017; 376(3): 221–234; doi: 10.1056/NEJMoa1601277.

54.  Montalban X, Hauser SL, Kappos L, et al. Ocrelizumab versus Placebo in Primary Progressive Multiple Sclerosis. N Engl J Med. 2017; 376(3): 209–220; doi: 10.1056/NEJMoa1606468.

55.  Lublin FD, Reingold SC, Cohen JA, et al. Defining the clinical course of multiple sclerosis: The 2013 revisions. Neurology. 2014; 83(3): 278–286; doi: 10.1212/WNL.0000000000000560.

56.  Coles AJ, Twyman CL, Arnold DL, et al. Alemtuzumab for patients with relapsing multiple sclerosis after disease-modifying therapy: a randomised controlled phase 3 trial. The Lancet. 2012; 380(9856): 1829–1839; doi: 10.1016/S0140-6736(12)61768-1.

57.  Jones JL, Anderson JM, Phuah C-L, et al. Improvement in disability after alemtuzumab treatment of multiple sclerosis is associated with neuroprotective autoimmunity. Brain. 2010; 133(8): 2232–2247.

58.  Furie R, Rovin BH, Houssiau F, et al. Two-Year, Randomized, Controlled Trial of Belimumab in Lupus Nephritis. N Engl J Med. 2020; 383(12): 1117–1128; doi: 10.1056/NEJMoa2001180.

59.  Chan AC, Carter PJ. Therapeutic antibodies for autoimmunity and inflammation. Nat Rev Immunol. 2010; 10(5): 301–316; doi: 10.1038/nri2761.

60.  Langley RG, Elewski BE, Lebwohl M, et al. Secukinumab in Plaque Psoriasis — Results of Two Phase 3 Trials. N Engl J Med. 2014; 371(4): 326–338; doi: 10.1056/NEJMoa1314258.

61.  Bai F, Li GG, Liu Q, et al. Short-Term Efficacy and Safety of IL-17, IL-12/23, and IL-23 Inhibitors Brodalumab, Secukinumab, Ixekizumab, Ustekinumab, Guselkumab, Tildrakizumab, and Risankizumab for the Treatment of Moderate to Severe Plaque Psoriasis: A Systematic Review and Network Meta-Analysis of Randomized Controlled Trials. Journal of Immunology Research. 2019; 2019: 1–25; doi: 10.1155/2019/2546161.

62.  Reich K, Armstrong AW, Langley RG, et al. Guselkumab versus secukinumab for the treatment of moderate-to-severe psoriasis (ECLIPSE): results from a phase 3, randomised controlled trial. The Lancet. 2019; 394(10201): 831–839.

63.  Malagoli P, Dapavo P, Amerio P, et al. Secukinumab in the Treatment of Psoriasis: A Narrative Review on Early Treatment and Real-World Evidence. Dermatol Ther (Heidelb). 2024; 14(10): 2739–2757; doi: 10.1007/s13555-024-01255-4.

64.  Gaikwad RG, Shinde AJ, Hajare AA. Herbal Treatment for Management of Psoriasis: An Overview. RJPT. 2022; 1385–1392; doi: 10.52711/0974-360X.2022.00231.

65.  Postow MA, Sidlow R, Hellmann MD. Immune-Related Adverse Events Associated with Immune Checkpoint Blockade. Longo DL. ed. N Engl J Med. 2018;378(2):158–168; doi: 10.1056/NEJMra1703481.

66.  Kazi DS, Moran AE, Coxson PG, et al. Cost-effectiveness of PCSK9 Inhibitor Therapy in Patients with Heterozygous Familial Hypercholesterolemia or Atherosclerotic Cardiovascular Disease. JAMA. 2016; 316(7):743; doi: 10.1001/jama.2016.11004.

67.  Feist E, Fatenejad S, Grishin S, et al. Olokizumab, a monoclonal antibody against interleukin-6, in combination with methotrexate in patients with rheumatoid arthritis inadequately controlled by tumour necrosis factor inhibitor therapy: efficacy and safety results of a randomised controlled phase III study. Annals of the Rheumatic Diseases. 2022; 81(12):1661–1668.

68.  Pisetsky DS, Rovin BH, Lipsky PE. New Perspectives in Rheumatology: Biomarkers as Entry Criteria for Clinical Trials of New Therapies for Systemic Lupus Erythematosus: The Example of Antinuclear Antibodies and Anti‐DNA. Arthritis and Rheumatology. 2017; 69(3): 487–493; doi: 10.1002/art.40008.

69.  Panwar B, Schmiedel BJ, Liang S, et al. Multi–cell type gene coexpression network analysis reveals coordinated interferon response and cross–cell type correlations in systemic lupus erythematosus. Genome Research. 2021; 31(4):659–676.

70.  Herbst R, Liu Z, Jallal B, et al. Biomarkers for systemic lupus erythematosus. Int J of Rheum Dis. 2012; 15(5): 433–444; doi: 10.1111/j.1756-185X.2012.01764.x.

71.  Tanaka Y, Kubo S, Miyagawa I, et al. Lymphocyte Phenotype and Its Application to Precision Medicine in Systemic Autoimmune Diseases. In: Seminars in Arthritis and Rheumatism Elsevier. 2019; pp. 1146–1150.

72.  Rezvani K, Rouce R, Liu E, et al. Engineering natural killer cells for cancer immunotherapy. Molecular Therapy. 2017; 25(8):1769–1781.

73.  Jeyakumar N, Smith M. Custom CARs: leveraging the adaptability of allogeneic CAR therapies to address current challenges in relapsed/refractory DLBCL. Frontiers in Immunology. 2022; 13: 887866.

74.  Robinson WH, Lindstrom TM, Cheung RK, et al. Mechanistic biomarkers for clinical decision making in rheumatic diseases. Nature Reviews Rheumatology.  2013; 9(5):267–276.

75.  Skougaard M, Drachmann C, Stisen ZR, et al. OP0103 Gene expression profiles retrieved from single cell rna sequencing reveal phenotypic traits in patients with psoriatic arthritis initiating tumour necrosis factor alpha inhibitor and interleukin-17 Inhibitor. 2022.

76.  Fischer JAA, Hueber AJ, Wilson S, et al. Combined Inhibition of Tumor Necrosis Factor α and Interleukin‐17 As a Therapeutic Opportunity in Rheumatoid Arthritis: Development and Characterization of a Novel Bispecific Antibody. Arthritis & Rheumatology.  2015; 67(1): 51–62; doi: 10.1002/art.38896.

77.  Biesemann N, Margerie D, Asbrand C, et al. Additive efficacy of a bispecific anti–TNF/IL-6 nanobody compound in translational models of rheumatoid arthritis. Sci Transl Med. 2023; 15(681): eabq4419; doi: 10.1126/scitranslmed.abq4419.

78.  Stadtmauer EA, Fraietta JA, Davis MM, et al. CRISPR-engineered T cells in patients with refractory cancer. Science. 2020; 367(6481): eaba7365; doi: 10.1126/science.aba7365.

79.  Fesnak AD, June CH, Levine BL. Engineered T cells: the promise and challenges of cancer immunotherapy. Nat Rev Cancer. 2016; 16(9): 566–581; doi: 10.1038/nrc.2016.97.

80.  Miotto R, Wang F, Wang S, et al. Deep learning for healthcare: review, opportunities and challenges. Briefings in Bioinformatics. 2018; 19(6): 1236–1246.

81.  Topol EJ. High-performance medicine: the convergence of human and artificial intelligence. Nat Med. 2019; 25(1):44–56; doi: 10.1038/s41591-018-0300-7.

82.  Bhatia T, Sharma S. Drug Repurposing: Insights into Current Advances and Future Applications. Current Medicinal Chemistry.  2024.

 

 

 

 

 

Received on 28.11.2024      Revised on 08.02.2025

Accepted on 17.03.2025      Published on 03.05.2025

Available online from May 05, 2025

Asian J. Pharm. Res. 2025; 15(2):183-191.

DOI: 10.52711/2231-5691.2025.00030

©Asian Pharma Press All Right Reserved

 

This work is licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International License. Creative Commons License.