Immune Checkpoint Inhibitors in Type 1 Diabetes: A New Frontier in Therapy

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/378744248

Immune Checkpoint Inhibitors in Type 1 Diabetes: A New Frontier in Therapy

Article · March 2024

CITATIONS READS
0 37

2 authors:

Emmanuel Ifeanyi Obeagu Getrude Uzoma Obeagu


Kampala International University (KIU) Kampala International University (KIU)
1,422 PUBLICATIONS 11,224 CITATIONS 384 PUBLICATIONS 3,307 CITATIONS

SEE PROFILE SEE PROFILE

All content following this page was uploaded by Emmanuel Ifeanyi Obeagu on 06 March 2024.

The user has requested enhancement of the downloaded file.


Elite Journal of Medicine. Volume 2 issue 2(2024), Pp. 26-41
https://epjournals.com/journals/EJM

Immune Checkpoint Inhibitors in Type 1 Diabetes: A New Frontier in Therapy

*Emmanuel Ifeanyi Obeagu1 and Getrude Uzoma Obeagu2

1Department of Medical Laboratory Science, Kampala International University, Uganda,


emmanuelobeagu@yahoo.com, ORCID: 0000-0002-4538-0161
2Schoolof Nursing Science, Kampala International University, Uganda,
uzomagertrude@gmail.com

*
Corresponding authour: Emmanuel Ifeanyi Obeagu, Department of Medical Laboratory Science,
Kampala International University, Uganda, emmanuelobeagu@yahoo.com, ORCID: 0000-0002-
4538-0161

Abstract

Type 1 Diabetes (T1D) is a chronic autoimmune disease characterized by the immune-mediated


destruction of insulin-producing beta cells in the pancreas. Current therapeutic strategies primarily
focus on glycemic control through exogenous insulin administration. However, emerging research
has explored the potential of immune checkpoint inhibitors (ICIs), initially developed for cancer
immunotherapy, as a novel therapeutic approach for T1D. This abstract provides a concise
overview of the current landscape of ICIs in T1D therapy, highlighting their mechanisms of action,
preclinical and clinical studies, challenges, and future prospects. ICIs target key immune
checkpoint molecules, including CTLA-4, PD-1, and PD-L1, aiming to modulate immune
responses. In T1D, these inhibitors hold the promise of reining in autoreactive T cells, thereby
preserving beta cell function and slowing disease progression. Extensive preclinical investigations
in murine models have demonstrated the efficacy of ICIs in preventing the autoimmune destruction
of beta cells. These studies provide crucial insights into the mechanisms underlying T-cell
regulation and offer a foundation for translating findings into clinical applications. In conclusion,
immune checkpoint inhibitors represent a promising frontier in T1D therapy, offering a paradigm
shift towards immune modulation for disease modification. This abstract provides a snapshot of
the current status of ICIs in T1D research and underscores the need for ongoing investigations,
precision medicine approaches, and collaborative efforts to unlock the full potential of these
innovative therapies.

Citation: Obeagu EI, Obeagu GU. Immune Checkpoint Inhibitors in Type 1 Diabetes: A New
Frontier in Therapy. Elite Journal of Medicine, 2024; 2(2): 26-41

1
Elite Journal of Medicine. Volume 2 issue 2(2024), Pp. 26-41
https://epjournals.com/journals/EJM

Keywords: Type 1 Diabetes, Immune Checkpoint Inhibitors, Autoimmunity, Immunotherapy, T-


cell Regulation, Beta Cell Preservation, Precision Medicine

Introduction

Type 1 Diabetes (T1D) is a chronic autoimmune disorder characterized by the selective destruction
of insulin-producing beta cells in the pancreas, resulting in a lifelong dependence on exogenous
insulin. Despite advancements in insulin therapy, the quest for disease-modifying treatments that
address the underlying autoimmune pathology remains a pressing need.1-4 In recent years, immune
checkpoint inhibitors (ICIs), originally designed to unleash the immune system against cancer
cells, have emerged as a novel and promising avenue for therapeutic intervention in T1D.5
Traditionally, T1D has been managed by insulin replacement therapy to control hyperglycemia
and prevent acute complications.6 However, this approach does not address the root cause of the
disease—the autoimmune assault on pancreatic beta cells. Immune checkpoint inhibitors (ICIs),
known for their success in cancer immunotherapy, operate by releasing the brakes on immune
responses, particularly by targeting key checkpoint molecules such as cytotoxic T-lymphocyte-
associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1), and its ligand PD-L1.
This innovative strategy holds the potential to modulate aberrant immune responses, preserving
beta cell function and slowing the progression of T1D.7

The aim of exploring immune checkpoint inhibitors (ICIs) in the context of Type 1 Diabetes (T1D)
is to investigate novel therapeutic strategies that could potentially modify the course of the disease.

Mechanisms of Action

Immune checkpoint inhibitors (ICIs) represent a class of therapeutic agents designed to modulate
the immune system by targeting specific checkpoint molecules involved in immune regulation.8
In the context of Type 1 Diabetes (T1D), these agents hold the potential to disrupt the autoimmune
attack on insulin-producing beta cells. Understanding the intricate mechanisms of action
underlying ICIs provides insights into their application and potential efficacy in T1D therapy.
Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) is a key inhibitory receptor expressed on
T cells.9 It competes with the co-stimulatory receptor CD28 for binding to B7 molecules on
antigen-presenting cells. By inhibiting T cell activation and proliferation, CTLA-4 acts as a crucial
checkpoint in immune homeostasis. ICIs targeting CTLA-4, such as ipilimumab, aim to unleash T
cell responses by blocking this inhibitory signal, potentially preventing the autoimmune
destruction of beta cells in T1D.10

The programmed cell death protein 1 (PD-1) receptor and its ligand PD-L1 play a pivotal role in
regulating T cell activity.11 In T1D, aberrant PD-1/PD-L1 interactions contribute to immune
tolerance breakdown.12 Immune checkpoint inhibitors (ICIs) like pembrolizumab and nivolumab
inhibit this interaction, allowing for sustained T cell activity. By disrupting this checkpoint, ICIs
may prevent the suppression of autoreactive T cells, limiting their assault on beta cells. Immune
checkpoint inhibitors (ICIs) exert their effects by modulating T-cell responses, either by enhancing
Citation: Obeagu EI, Obeagu GU. Immune Checkpoint Inhibitors in Type 1 Diabetes: A New
Frontier in Therapy. Elite Journal of Medicine, 2024; 2(2): 26-41

2
Elite Journal of Medicine. Volume 2 issue 2(2024), Pp. 26-41
https://epjournals.com/journals/EJM

effector T-cell function or by inhibiting regulatory T-cell activity.13 In the context of T1D, the
delicate balance between effector and regulatory T cells is disrupted, leading to autoimmune
destruction. Immune checkpoint inhibitors (ICIs) aim to recalibrate this balance, promoting
immune tolerance and mitigating the autoimmune attack on beta cells.

Immune checkpoint inhibitors (ICIs) have the potential to enhance antigen-specific immune
responses.14 In T1D, the identification and targeting of specific autoantigens associated with beta
cells can be critical. By unleashing the immune system's specificity, ICIs may redirect T-cell
responses towards antigens that promote regulatory mechanisms, thereby attenuating the
autoimmune response against beta cells. Beyond the specific targeting of checkpoint molecules,
Immune checkpoint inhibitors (ICIs) induce a broader immunomodulatory effect. This includes
the activation of various immune cell types, changes in cytokine profiles, and alterations in the
tumor microenvironment. In T1D, such immunomodulation may lead to a shift from a pro-
inflammatory to an anti-inflammatory state, promoting beta cell survival.15-16

Preclinical Studies

Preclinical studies serve as a critical foundation for assessing the efficacy, safety, and mechanisms
of action of immune checkpoint inhibitors (ICIs) in the context of Type 1 Diabetes (T1D).17-18
These investigations, often conducted in murine models of T1D, offer valuable insights into the
potential of ICIs for preserving beta cell function and altering the autoimmune trajectory of the
disease. Preclinical studies frequently employ murine models, such as non-obese diabetic (NOD)
mice, that share immunological and genetic features with human T1D.19-20 These models allow
researchers to recapitulate the autoimmune destruction of beta cells, providing a relevant platform
to evaluate the impact of ICIs on disease progression.

Numerous preclinical studies have demonstrated the efficacy of ICIs in preserving beta cell mass
and function.21-22 For example, anti-CTLA-4 antibodies and anti-PD-1/PD-L1 antibodies have
shown the ability to attenuate the infiltration of autoreactive T cells into pancreatic islets, reducing
the severity of insulitis and maintaining insulin-producing capacity. Preclinical investigations
focus on deciphering the specific effects of ICIs on immune responses in T1D.23-24 These studies
often reveal a modulation of T-cell activity, a shift in cytokine profiles, and changes in the balance
between effector and regulatory T cells. The goal is to establish a more tolerogenic immune
environment that protects beta cells from autoimmune attack.

Markers of autoimmune activity, such as autoantibodies and cytokine levels, are carefully assessed
in preclinical studies.25-26 Changes in these biomarkers provide insights into the direct impact of
ICIs on the autoimmune processes associated with T1D, helping to identify potential surrogate
markers for treatment response. Preclinical studies contribute to determining optimal dosages of
ICIs and assessing their safety profiles. Understanding the dose-response relationship is crucial for
translating preclinical findings to clinical settings. Additionally, these studies help identify
potential adverse effects and inform strategies for minimizing off-target effects. Exploration of
combination therapies is a key aspect of preclinical research. Combinations of ICIs with other
Citation: Obeagu EI, Obeagu GU. Immune Checkpoint Inhibitors in Type 1 Diabetes: A New
Frontier in Therapy. Elite Journal of Medicine, 2024; 2(2): 26-41

3
Elite Journal of Medicine. Volume 2 issue 2(2024), Pp. 26-41
https://epjournals.com/journals/EJM

immunomodulatory agents or standard T1D treatments are investigated to enhance therapeutic


efficacy while minimizing potential side effects.27-28 The synergistic effects of such combinations
are evaluated in preclinical models. The insights gained from preclinical studies guide the design
of clinical trials and inform translational approaches. These studies are instrumental in shaping the
rationale for moving ICIs from bench to bedside, providing a basis for further investigations in
human subjects.

Clinical Trials

Clinical trials represent a crucial phase in the translation of immune checkpoint inhibitors (ICIs)
from preclinical promise to tangible therapeutic interventions for Type 1 Diabetes (T1D) patients.
These trials aim to evaluate the safety, efficacy, and tolerability of ICIs in a human population,
providing essential insights into their potential as disease-modifying agents. Clinical trials
assessing ICIs in T1D carefully design protocols to investigate specific objectives. Patient
selection criteria consider factors such as disease duration, age, and baseline insulin requirements.
Additionally, inclusion criteria often involve assessing autoantibody status to identify individuals
at different stages of autoimmune progression.29-30 Clinical trials establish primary and secondary
endpoints to measure the therapeutic impact of ICIs.31-32 Primary endpoints may include
preservation of beta cell function, reduction in insulin requirements, or improvement in glycemic
control. Secondary endpoints often encompass immunological markers, quality of life measures,
and safety assessments.

Several clinical trials have investigated the efficacy of anti-CTLA-4 antibodies (e.g., ipilimumab)
and anti-PD-1/PD-L1 antibodies (e.g., pembrolizumab, nivolumab) in T1D.33-34 These trials
explore the ability of ICIs to modulate autoimmune responses, with a particular focus on
preserving beta cell function and reducing the need for exogenous insulin. Clinical trials involve
longitudinal assessments to capture the dynamic changes in disease progression and treatment
response. Regular monitoring of glycemic parameters, autoimmune biomarkers, and safety
profiles over an extended period allows for a comprehensive evaluation of the sustained effects of
ICIs. Advancements in precision medicine are reflected in clinical trials, where efforts are made
to identify biomarkers predictive of treatment response.35-36 Stratifying patients based on their
immunological profiles or genetic predispositions enables a more personalized approach,
optimizing the likelihood of positive outcomes.

Clinical trials meticulously monitor and report safety data associated with ICIs. Adverse events,
including immune-related adverse events (irAEs), are systematically documented to assess the
risk-benefit profile. This information guides clinicians in managing potential side effects and
informs future trial designs.37-38 Clinical trials typically progress through different phases. Phase I
trials establish safety and dosing, Phase II trials assess efficacy and optimal dosages, and Phase III
trials involve larger populations to confirm efficacy and monitor long-term safety. The progression
from phase to phase is contingent on positive results and a favorable risk-benefit ratio. Building
on insights from preclinical studies, clinical trials explore combination therapies involving ICIs

Citation: Obeagu EI, Obeagu GU. Immune Checkpoint Inhibitors in Type 1 Diabetes: A New
Frontier in Therapy. Elite Journal of Medicine, 2024; 2(2): 26-41

4
Elite Journal of Medicine. Volume 2 issue 2(2024), Pp. 26-41
https://epjournals.com/journals/EJM

and other immunomodulatory agents.39 This approach aims to enhance therapeutic outcomes by
targeting multiple facets of the autoimmune response.

Challenges and Considerations

The exploration of immune checkpoint inhibitors (ICIs) as a therapeutic strategy for Type 1
Diabetes (T1D) is met with several challenges and considerations. While promising, these
challenges need to be carefully addressed to ensure the safety, efficacy, and feasibility of ICI-
based therapies in the complex landscape of autoimmune diabetes. Type 1 Diabetes (T1D) is a
heterogeneous disease with variability in disease onset, progression, and underlying
immunopathology.40 Patient heterogeneity poses a challenge in identifying a uniform response to
ICIs. Tailoring therapies to individualized immune profiles and disease stages is crucial for
optimizing treatment outcomes. Immune checkpoint inhibitors (ICIs) have demonstrated success
in cancer immunotherapy but are associated with immune-related adverse events (irAEs).41 The
long-term safety of these agents in the context of T1D, especially considering the chronic nature
of the disease, raises concerns. Continuous monitoring for potential side effects and the
development of strategies to mitigate irAEs are essential.

Determining the optimal timing for ICI intervention is challenging. Type 1 Diabetes (T1D) is often
diagnosed after significant beta cell loss has occurred.42 Early intervention may be more effective
in preserving beta cell function, but the identification of at-risk individuals before clinical onset
presents a considerable challenge. Immune checkpoint inhibitors (ICIs) aim to modulate immune
responses, but achieving a delicate balance between suppressing autoimmune attacks and
maintaining protective immunity is challenging.43 Indiscriminate immune activation may lead to
unintended consequences, including off-target effects and the risk of exacerbating autoimmunity.
The identification of reliable biomarkers predictive of treatment response remains a significant
challenge. Establishing biomarkers that correlate with disease progression, treatment efficacy, and
potential side effects is crucial for patient stratification and personalized medicine approaches.

Developing individualized treatment strategies based on the unique immunological profiles of T1D
patients is a complex task.44 Factors such as age, genetic predisposition, and the presence of
specific autoantibodies may influence treatment response, necessitating a personalized medicine
approach. While combination therapies hold promise, understanding the synergistic effects and
potential synergistic toxicities of combining ICIs with other agents is challenging. Identifying the
right combination partners and optimizing dosages require careful consideration to maximize
therapeutic benefits. Patient adherence to treatment regimens, especially if involving repeated
administrations of ICIs, is critical for therapeutic success.45 Additionally, patient acceptance of
potential risks and benefits, as well as their understanding of the chronic nature of T1D, is essential
for the successful implementation of ICI-based therapies.

Future Directions

Citation: Obeagu EI, Obeagu GU. Immune Checkpoint Inhibitors in Type 1 Diabetes: A New
Frontier in Therapy. Elite Journal of Medicine, 2024; 2(2): 26-41

5
Elite Journal of Medicine. Volume 2 issue 2(2024), Pp. 26-41
https://epjournals.com/journals/EJM

The exploration of immune checkpoint inhibitors (ICIs) in Type 1 Diabetes (T1D) therapy has
opened new avenues for disease modification. As research progresses, several future directions
emerge, presenting opportunities to enhance the efficacy, safety, and applicability of ICI-based
approaches. Advancements in precision medicine hold the key to tailoring ICI therapies to the
unique immunological profiles of T1D patients.46 Identifying specific biomarkers predictive of
treatment response and disease progression will enable more targeted and personalized
interventions. Efforts to intervene at earlier stages of T1D, potentially in at-risk individuals before
clinical onset, could enhance the efficacy of ICI therapies.47 Developing reliable markers for early
disease detection and establishing the feasibility of preventive interventions are crucial future
directions. Exploring synergistic combinations of ICIs with other immunomodulatory agents or
standard T1D treatments is a promising avenue.48 Identifying complementary mechanisms of
action and optimizing dosages will be essential for maximizing therapeutic benefits while
minimizing potential side effects.

Continued research into the development of next-generation checkpoint inhibitors with improved
specificity and reduced off-target effects is crucial. Engineering molecules that selectively
modulate autoimmune responses without compromising protective immunity is a key focus for
future drug development. Dedicated efforts to discover and validate robust biomarkers for T1D
progression and treatment response are imperative.49 These biomarkers will not only aid in patient
stratification but also serve as critical tools for monitoring disease activity and treatment efficacy.
T1D often manifests in childhood, and considering the unique immunological aspects of pediatric
populations is essential.50 Future research should focus on understanding the dynamics of immune
responses in pediatric T1D and optimizing ICI therapies for this specific demographic. Continued
exploration of the mechanistic aspects of ICIs in the context of T1D is essential. Unraveling the
intricacies of how these agents modulate immune responses, influence regulatory T cells, and
impact the autoimmune milieu will provide valuable insights for refining therapeutic strategies.

Adapting regulatory frameworks to accommodate the unique considerations of autoimmune


diseases, including T1D, is critical.51 Future directions involve engaging regulatory agencies to
streamline approval processes and address the specific challenges associated with ICI-based
therapies for T1D. Incorporating patient-reported outcomes in clinical trials and real-world settings
is crucial for understanding the holistic impact of ICI therapies on the lives of individuals with
T1D.52-54 This includes assessing quality of life, treatment satisfaction, and the psychological
aspects of managing a chronic autoimmune condition. Encouraging global collaborations and data-
sharing initiatives will facilitate a more comprehensive understanding of the diverse genetic and
environmental factors influencing T1D.55 Large-scale collaborative efforts can accelerate progress
and enhance the generalizability of research findings.

Implications for Clinical and Health Policy Making

The exploration of immune checkpoint inhibitors (ICIs) in Type 1 Diabetes (T1D) represents a
potential paradigm shift in therapeutic approaches. Clinically, this necessitates a shift from
primarily insulin-focused management to immune-modulating strategies aimed at disease
Citation: Obeagu EI, Obeagu GU. Immune Checkpoint Inhibitors in Type 1 Diabetes: A New
Frontier in Therapy. Elite Journal of Medicine, 2024; 2(2): 26-41

6
Elite Journal of Medicine. Volume 2 issue 2(2024), Pp. 26-41
https://epjournals.com/journals/EJM

modification. Health policies should encourage and support the integration of precision medicine
approaches into clinical practice. Identifying specific biomarkers predictive of treatment response
will enable tailored, patient-specific interventions, optimizing the use of ICIs in T1D. Clinical
guidelines and policies should be updated to consider and endorse early intervention strategies,
aiming to preserve beta cell function and prevent disease progression. This may involve screening
programs for at-risk individuals and the inclusion of early intervention protocols in standard care.
As evidence accumulates, health policies should consider incorporating ICIs into T1D treatment
guidelines. Clear recommendations on the use of ICIs, including patient selection criteria and
optimal timing of intervention, will guide clinicians in implementing these novel therapies.56-67

Health policies should establish robust safety monitoring protocols for ICI-based therapies in T1D.
Clear guidelines for the identification, management, and reporting of immune-related adverse
events (irAEs) will ensure the safe implementation of these therapies in clinical settings.
Regulatory bodies should adapt existing frameworks to accommodate the unique considerations
of autoimmune diseases, specifically T1D. Streamlining approval processes for ICIs in T1D and
addressing regulatory challenges will facilitate timely access to these innovative therapies. Health
policies should conduct cost-effectiveness assessments of ICI-based therapies for T1D. These
assessments will inform reimbursement decisions, ensuring that these novel treatments are
economically viable and accessible to a broad patient population. Health policies should support
education and training programs for healthcare professionals to enhance their understanding of the
immunological basis of T1D and the potential role of ICIs. This ensures that clinicians are well-
equipped to integrate these therapies into their practice. Policies should address issues of patient
access and equity to prevent disparities in the adoption of ICI-based therapies. This may involve
considering reimbursement policies, ensuring affordability, and promoting inclusive participation
in clinical trials.68-87

Health policies should support public awareness campaigns to inform individuals with T1D, their
families, and the broader community about the potential of ICIs. Educating the public will foster
informed decision-making and reduce stigma associated with novel therapies. Health policies
should encourage international collaboration to facilitate data-sharing initiatives, harmonize
regulatory standards, and promote global research efforts. Collaborative endeavors enhance the
robustness and generalizability of findings, benefiting the global T1D community. Given the rapid
pace of scientific advancements, health policies should adopt adaptive frameworks that can
respond to emerging evidence. This flexibility ensures that policies remain dynamic,
accommodating new knowledge and innovations in the field of T1D management.88-94

Recommendations

Foster collaborations among immunologists, endocrinologists, geneticists, and other specialists to


leverage diverse expertise in advancing research and clinical applications of immune checkpoint
inhibitors (ICIs) for Type 1 Diabetes (T1D). Invest in precision medicine initiatives to identify and
validate biomarkers that can guide patient stratification, predict treatment responses, and support
the development of personalized ICI-based therapeutic strategies. Prioritize research focused on
Citation: Obeagu EI, Obeagu GU. Immune Checkpoint Inhibitors in Type 1 Diabetes: A New
Frontier in Therapy. Elite Journal of Medicine, 2024; 2(2): 26-41

7
Elite Journal of Medicine. Volume 2 issue 2(2024), Pp. 26-41
https://epjournals.com/journals/EJM

understanding the unique immunological aspects of pediatric-onset T1D and tailor ICI therapies
to address the specific needs and challenges of younger populations. Design and conduct clinical
trials that explore the feasibility and efficacy of early intervention strategies, aiming to preserve
beta cell function and prevent the progression of T1D in at-risk individuals before clinical onset.

Support longitudinal studies to assess the long-term safety and efficacy of ICI-based therapies in
T1D, capturing the dynamic nature of autoimmune responses and treatment effects over extended
periods. Invest in the research and development of next-generation ICIs with improved specificity,
reduced off-target effects, and enhanced efficacy. This includes exploring novel molecular targets
and engineering approaches. Encourage research into synergistic combination therapies involving
ICIs and other immunomodulatory agents or standard T1D treatments. Optimize dosages and
assess the potential for enhanced therapeutic benefits while minimizing adverse effects. Integrate
patient-reported outcomes into clinical trials and real-world studies to capture the holistic impact
of ICI therapies on individuals with T1D. Assess quality of life, treatment satisfaction, and
psychological well-being to inform patient-centered care.

Engage with regulatory bodies to adapt frameworks for autoimmune diseases, facilitating the
efficient and safe approval of ICI-based therapies for T1D. Advocate for tailored regulatory
approaches that consider the unique characteristics of autoimmune conditions. Promote global
collaboration and data-sharing initiatives to create a comprehensive understanding of the genetic,
environmental, and immunological factors influencing T1D. Large-scale collaborations can
accelerate progress and enhance the generalizability of research findings. Invest in public
awareness and education campaigns to inform individuals with T1D, their families, and healthcare
professionals about the potential of ICIs as a novel therapeutic approach. Enhance understanding
of the evolving landscape of T1D management. Ensure inclusivity in clinical trial enrollment by
considering diverse demographics, including different age groups, ethnicities, and disease stages.
This inclusivity enhances the generalizability of trial results to a broader T1D population.

Conclusion

The exploration of immune checkpoint inhibitors (ICIs) in the context of Type 1 Diabetes (T1D)
marks a transformative chapter in the pursuit of disease-modifying therapies. The intricate
interplay between the immune system and beta cells has led researchers and clinicians to
investigate novel strategies, and ICIs represent a promising frontier in T1D management. As
research progresses, the implications for clinical practice and health policy making are profound.
The potential for a therapeutic paradigm shift, embracing precision medicine, and redefining early
intervention strategies all underscore the transformative impact of ICIs. While challenges exist,
from patient heterogeneity to long-term safety considerations, addressing these hurdles presents
opportunities for innovation and progress.

References
1. Obeagu EI, Obeagu GU. Type 1 diabetes mellitus: Roles of neutrophils in the pathogenesis.
Medicine. 2023;102(50): e36245.
Citation: Obeagu EI, Obeagu GU. Immune Checkpoint Inhibitors in Type 1 Diabetes: A New
Frontier in Therapy. Elite Journal of Medicine, 2024; 2(2): 26-41

8
Elite Journal of Medicine. Volume 2 issue 2(2024), Pp. 26-41
https://epjournals.com/journals/EJM

2. Okoroiwu IL, Obeagu EI, San Miguel HG, Bote SA, Obeagu GU. Characterisation of
HLA-DR antigen in patients type 1 diabetes mellitus in patient attending a tertairy hospital
in Enugu, south-east Nigeria. ACADEMIC JOURNAL. 2023.
3. Obeagu EI, Okoroiwu IL, Obeagu GU. Some haematological variables in insulin
dependent diabetes mellitus patients in Imo state Nigeria. Int. J. Curr. Res. Chem. Pharm.
Sci. 2016;3(4):110-7.
4. Okoroiwu IL, Obeagu EI, Obeagu GU, Chikezie CC, Ezema GO. The prevalence of
selected autoimmune diseases. Int. J. Adv. Multidiscip. Res. 2016;3(3):9-14.
5. Kiaie SH, Salehi-Shadkami H, Sanaei MJ, Azizi M, Shokrollahi Barough M, Nasr MS,
Sheibani M. Nano-immunotherapy: overcoming delivery challenge of immune checkpoint
therapy. Journal of nanobiotechnology. 2023;21(1):339.
6. Janež A, Guja C, Mitrakou A, Lalic N, Tankova T, Czupryniak L, Tabák AG, Prazny M,
Martinka E, Smircic-Duvnjak L. Insulin therapy in adults with type 1 diabetes mellitus: a
narrative review. Diabetes Therapy. 2020:387-409.
7. Shiravand Y, Khodadadi F, Kashani SM, Hosseini-Fard SR, Hosseini S, Sadeghirad H,
Ladwa R, O’Byrne K, Kulasinghe A. Immune checkpoint inhibitors in cancer therapy.
Current Oncology. 2022;29(5):3044-3060.
8. Singh S, Hassan D, Aldawsari HM, Molugulu N, Shukla R, Kesharwani P. Immune
checkpoint inhibitors: a promising anticancer therapy. Drug discovery today.
2020;25(1):223-9.
9. Azimnasab-Sorkhabi P, Soltani-Asl M, Kfoury Junior JR. Cytotoxic T-lymphocyte-
associated protein 4 (CTLA-4) as an undetermined tool in tumor cells. Human Cell.
2023:1-8.
10. Kuske M, Haist M, Jung T, Grabbe S, Bros M. Immunomodulatory properties of immune
checkpoint inhibitors—more than boosting T-cell responses? Cancers. 2022;14(7):1710.
11. Constantinidou A, Alifieris C, Trafalis DT. Targeting programmed cell death-1 (PD-1) and
ligand (PD-L1): a new era in cancer active immunotherapy. Pharmacology & Therapeutics.
2019; 194:84-106.
12. Fife BT, Pauken KE. The role of the PD‐1 pathway in autoimmunity and peripheral
tolerance. Annals of the New York Academy of Sciences. 2011;1217(1):45-59.
13. Marei HE, Hasan A, Pozzoli G, Cenciarelli C. Cancer immunotherapy with immune
checkpoint inhibitors (ICIs): potential, mechanisms of resistance, and strategies for
reinvigorating T cell responsiveness when resistance is acquired. Cancer Cell International.
2023;23(1):64.
14. Lee J, Kim EH. Mechanisms underlying response and resistance to immune checkpoint
blockade in cancer immunotherapy. Frontiers in Oncology. 2023;13.
15. Varayathu H, Sarathy V, Thomas BE, Mufti SS, Naik R. Combination strategies to
augment immune check point inhibitors efficacy-implications for translational research.
Frontiers in oncology. 2021; 11:559161.
16. Simpson RC, Shanahan ER, Scolyer RA, Long GV. Towards modulating the gut
microbiota to enhance the efficacy of immune-checkpoint inhibitors. Nature Reviews
Clinical Oncology. 2023;20(10):697-715.

Citation: Obeagu EI, Obeagu GU. Immune Checkpoint Inhibitors in Type 1 Diabetes: A New
Frontier in Therapy. Elite Journal of Medicine, 2024; 2(2): 26-41

9
Elite Journal of Medicine. Volume 2 issue 2(2024), Pp. 26-41
https://epjournals.com/journals/EJM

17. Cantley J, Eizirik DL, Latres E, Dayan CM. Islet cells in human type 1 diabetes: from
recent advances to novel therapies–a symposium-based roadmap for future research.
Journal of Endocrinology. 2023;259(1).
18. Gacche RN. Changing landscape of anti-angiogenic therapy: Novel approaches and clinical
perspectives. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer. 2023:189020.
19. Pearson JA, Wong FS, Wen L. The importance of the Non Obese Diabetic (NOD) mouse
model in autoimmune diabetes. Journal of autoimmunity. 2016; 66:76-88.
20. Funda DP, Palová-Jelínková L, Goliáš J, Kroulíková Z, Fajstová A, Hudcovic T, Špíšek R.
Optimal tolerogenic dendritic cells in type 1 diabetes (T1D) therapy: what can we learn
from non-obese diabetic (NOD) mouse models? Frontiers in Immunology. 2019; 10:967.
21. Chow A, Perica K, Klebanoff CA, Wolchok JD. Clinical implications of T cell exhaustion
for cancer immunotherapy. Nature reviews Clinical oncology. 2022;19(12):775-90.
22. Hayase E, Jenq RR. Role of the intestinal microbiome and microbial-derived metabolites
in immune checkpoint blockade immunotherapy of cancer. Genome medicine.
2021;13(1):107.
23. Cina ML, Venegas J, Young A. Stocking the toolbox—Using preclinical models to
understand the development and treatment of immune checkpoint inhibitor‐induced
immune‐related adverse events. Immunological Reviews. 2023;318(1):110-37.
24. Calabrese LH. Immunopathogenesis of Immune-Related Adverse Events from Cancer
Immunotherapy. Rheumatic Diseases and Syndromes Induced by Cancer Immunotherapy:
A Handbook for Diagnosis and Management. 2021:49-68.
25. Ma WT, Chang C, Gershwin ME, Lian ZX. Development of autoantibodies precedes
clinical manifestations of autoimmune diseases: A comprehensive review. Journal of
autoimmunity. 2017; 83:95-112.
26. Kalinkovich A, Gabdulina G, Livshits G. Autoimmunity, inflammation, and dysbiosis
mutually govern the transition from the preclinical to the clinical stage of rheumatoid
arthritis. Immunologic Research. 2018; 66:696-709.
27. Isaacs D, Drakaki A, Xing Y, In G, Angell T, Lechner M, Hilder R, Tsai K, Quandt Z.
Safety and efficacy of immune checkpoint inhibitor cancer therapy in patients with
preexisting type 1 diabetes mellitus.
28. Ding JT, Yang KP, Lin KL, Cao YK, Zou F. Mechanisms and therapeutic strategies of
immune checkpoint molecules and regulators in type 1 diabetes. Frontiers in
Endocrinology. 2023; 13:1090842.
29. Ji HH, Tang XW, Dong Z, Song L, Jia YT. Adverse event profiles of anti-CTLA-4 and
anti-PD-1 monoclonal antibodies alone or in combination: analysis of spontaneous reports
submitted to FAERS. Clinical drug investigation. 2019; 39:319-30.
30. Melaiu O, Lucarini V, Giovannoni R, Fruci D, Gemignani F. News on immune checkpoint
inhibitors as immunotherapy strategies in adult and pediatric solid tumors. InSeminars in
cancer biology 2022; 79:18-43. Academic Press.
31. Reynolds KL, Arora S, Elayavilli RK, Louv WC, Schaller TH, Khandelwal A, Rothenberg
M, Khozin S, Guidon AC, Dougan M, Zubiri L. Immune-related adverse events associated
with immune checkpoint inhibitors: a call to action for collecting and sharing clinical trial
and real-world data. Journal for ImmunoTherapy of Cancer. 2021;9(7).
Citation: Obeagu EI, Obeagu GU. Immune Checkpoint Inhibitors in Type 1 Diabetes: A New
Frontier in Therapy. Elite Journal of Medicine, 2024; 2(2): 26-41

10
Elite Journal of Medicine. Volume 2 issue 2(2024), Pp. 26-41
https://epjournals.com/journals/EJM

32. Bagchi S, Yuan R, Engleman EG. Immune checkpoint inhibitors for the treatment of
cancer: clinical impact and mechanisms of response and resistance. Annual Review of
Pathology: Mechanisms of Disease. 2021; 16:223-249.
33. Wojtukiewicz MZ, Rek MM, Karpowicz K, Górska M, Polityńska B, Wojtukiewicz AM,
Moniuszko M, Radziwon P, Tucker SC, Honn KV. Inhibitors of immune checkpoints—
PD-1, PD-L1, CTLA-4—new opportunities for cancer patients and a new challenge for
internists and general practitioners. Cancer and Metastasis Reviews. 2021; 40:949-982.
34. Wong SK, Beckermann KE, Johnson DB, Das S. Combining anti-cytotoxic T-lymphocyte
antigen 4 (CTLA-4) and-programmed cell death protein 1 (PD-1) agents for cancer
immunotherapy. Expert Opinion on Biological Therapy. 2021;21(12):1623-1634.
35. Seyhan AA, Carini C. Are innovation and new technologies in precision medicine paving
a new era in patients centric care? Journal of translational medicine. 2019; 17:1-28.
36. Tarhini A, Kudchadkar RR. Predictive and on-treatment monitoring biomarkers in
advanced melanoma: Moving toward personalized medicine. Cancer treatment reviews.
2018; 71:8-18.
37. Rzeniewicz K, Larkin J, Menzies AM, Turajlic S. Immunotherapy use outside clinical trial
populations: never say never? Annals of Oncology. 2021;32(7):866-880.
38. Pasello G, Pavan A, Attili I, Bortolami A, Bonanno L, Menis J, Conte P, Guarneri V. Real
world data in the era of Immune Checkpoint Inhibitors (ICIs): Increasing evidence and
future applications in lung cancer. Cancer treatment reviews. 2020; 87:102031.
39. Fulgenzi CA, D’Alessio A, Ogunbiyi O, Demirtas CO, Gennari A, Cortellini A, Sharma
R, Pinato DJ. Novel immunotherapy combinations in clinical trials for hepatocellular
carcinoma: will they shape the future treatment landscape? Expert Opinion on
Investigational Drugs. 2022;31(7):681-691.
40. Infante M, Alejandro R, Fabbri A, Ricordi C. The heterogeneity of type 1 diabetes: From
immunopathology to immune intervention. InTranslational Autoimmunity 2022: 83-104.
Academic Press.
41. Lee DJ, Lee HJ, Farmer JR, Reynolds KL. Mechanisms driving immune-related adverse
events in cancer patients treated with immune checkpoint inhibitors. Current Cardiology
Reports. 2021; 23:1-2.
42. Oram RA, Sims EK, Evans-Molina C. Beta cells in type 1 diabetes: mass and function;
sleeping or dead? Diabetologia. 2019; 62:567-77.
43. Jia Y, Liu L, Shan B. Future of immune checkpoint inhibitors: Focus on tumor immune
microenvironment. Annals of Translational Medicine. 2020;8(17).
44. Linsley PS, Greenbaum CJ, Nepom GT. Uncovering pathways to personalized therapies in
type 1 diabetes. Diabetes. 2021;70(4):831-41.
45. Huang A, Yang XR, Chung WY, Dennison AR, Zhou J. Targeted therapy for
hepatocellular carcinoma. Signal transduction and targeted therapy. 2020;5(1):146.
46. Deligiorgi MV, Trafalis DT. A Concerted Vision to Advance the Knowledge of Diabetes
Mellitus Related to Immune Checkpoint Inhibitors. International Journal of Molecular
Sciences. 2023;24(8):7630.

Citation: Obeagu EI, Obeagu GU. Immune Checkpoint Inhibitors in Type 1 Diabetes: A New
Frontier in Therapy. Elite Journal of Medicine, 2024; 2(2): 26-41

11
Elite Journal of Medicine. Volume 2 issue 2(2024), Pp. 26-41
https://epjournals.com/journals/EJM

47. Lu J, Yang J, Liang Y, Meng H, Zhao J, Zhang X. Incidence of immune checkpoint


inhibitor-associated diabetes: a meta-analysis of randomized controlled studies. Frontiers
in pharmacology. 2019; 10:1453.
48. Kiaie SH, Salehi-Shadkami H, Sanaei MJ, Azizi M, Shokrollahi Barough M, Nasr MS,
Sheibani M. Nano-immunotherapy: overcoming delivery challenge of immune checkpoint
therapy. Journal of nanobiotechnology. 2023;21(1):339.
49. Yi L, Swensen AC, Qian WJ. Serum biomarkers for diagnosis and prediction of type 1
diabetes. Translational Research. 2018; 201:13-25.
50. Bauer W, Gyenesei A, Krętowski A. The multifactorial progression from the islet
autoimmunity to type 1 diabetes in children. International Journal of Molecular Sciences.
2021;22(14):7493.
51. James EA, Joglekar AV, Linnemann AK, Russ HA, Kent SC. The beta cell-immune cell
interface in type 1 diabetes (T1D). Molecular Metabolism. 2023:101809.
52. Bayless NL, Bluestone JA, Bucktrout S, Butterfield LH, Jaffee EM, Koch CA, Roep BO,
Sharpe AH, Murphy WJ, Villani AC, Walunas TL. Development of preclinical and clinical
models for immune-related adverse events following checkpoint immunotherapy: a
perspective from SITC and AACR. Journal for immunotherapy of cancer. 2021;9(9).
53. Emens LA, Adams S, Cimino-Mathews A, Disis ML, Gatti-Mays ME, Ho AY, Kalinsky
K, McArthur HL, Mittendorf EA, Nanda R, Page DB. Society for Immunotherapy of
Cancer (SITC) clinical practice guideline on immunotherapy for the treatment of breast
cancer. Journal for immunotherapy of cancer. 2021;9(8).
54. Gusev A. Germline mechanisms of immunotherapy toxicities in the era of genome‐wide
association studies. Immunological Reviews. 2023;318(1):138-156.
55. Flannick J, Florez JC. Type 2 diabetes: genetic data sharing to advance complex disease
research. Nature Reviews Genetics. 2016;17(9):535-549.
56. Gacche RN. Changing landscape of anti-angiogenic therapy: Novel approaches and clinical
perspectives. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer. 2023:189020.
57. Pritchard DE, Moeckel F, Villa MS, Housman LT, McCarty CA, McLeod HL. Strategies
for integrating personalized medicine into healthcare practice. Personalized medicine.
2017;14(2):141-52.
58. Piepoli MF, Corrà U, Adamopoulos S, Benzer W, Bjarnason-Wehrens B, Cupples M,
Dendale P, Doherty P, Gaita D, Höfer S, McGee H. Secondary prevention in the clinical
management of patients with cardiovascular diseases. Core components, standards and
outcome measures for referral and delivery: a policy statement from the cardiac
rehabilitation section of the European Association for Cardiovascular Prevention &
Rehabilitation. Endorsed by the Committee for Practice Guidelines of the European
Society of Cardiology. European journal of preventive cardiology. 2014;21(6):664-81.
59. Richardson SJ, Pugliese A. 100 YEARS OF INSULIN: Pancreas pathology in type 1
diabetes: an evolving story. Journal of endocrinology. 2022;252(2): R41-57.
60. Wright JJ, Powers AC, Johnson DB. Endocrine toxicities of immune checkpoint inhibitors.
Nature Reviews Endocrinology. 2021;17(7):389-99.
61. Ross ER, Altimus C. Type 1 Diabetes Autoantibody Screening: A Roadmap for Pediatric
Policy Implementation. Milken Institute. 2021.
Citation: Obeagu EI, Obeagu GU. Immune Checkpoint Inhibitors in Type 1 Diabetes: A New
Frontier in Therapy. Elite Journal of Medicine, 2024; 2(2): 26-41

12
Elite Journal of Medicine. Volume 2 issue 2(2024), Pp. 26-41
https://epjournals.com/journals/EJM

62. Von Itzstein MS, Gonugunta AS, Mayo HG, Minna JD, Gerber DE. Immunotherapy use
in patients with lung cancer and comorbidities. Cancer journal (Sudbury, Mass.).
2020;26(6):525.
63. Akturk HK, Couts KL, Baschal EE, Karakus KE, Van Gulick RJ, Turner JA, Pyle L,
Robinson WA, Michels AW. Analysis of human leukocyte antigen DR alleles, immune-
related adverse events, and survival associated with immune checkpoint inhibitor use
among patients with advanced malignant melanoma. JAMA Network Open.
2022;5(12):e2246400-.
64. Mountzios G, Remon J, Hendriks LE, García-Campelo R, Rolfo C, Van Schil P, Forde
PM, Besse B, Subbiah V, Reck M, Soria JC. Immune-checkpoint inhibition for resectable
non-small-cell lung cancer—Opportunities and challenges. Nature Reviews Clinical
Oncology. 2023;20(10):664-77.
65. Hedman Ahlström B. Focus on Every-day Life: Internet-based Support and Coaching for
Young Adults with Neuropsychiatric Disorders-A Chat-log Analysis. InIV. World
Congress on Social Media and Web 2.0 in Medicine, Health and Biomedical research.
Medicine 2.0 at Stanford university 2011: 27-28.
66. Núñez-Baila MÁ, Gómez-Aragón A, Marques-Silva AM, González-López JR. Lifestyle in
Emerging Adults with Type 1 Diabetes Mellitus: A Qualitative Systematic Review.
InHealthcare 2024; 12(3):309. MDPI.
67. Phiri T, Mowat R, Cook C. What nursing interventions and healthcare practices facilitate
type 1 diabetes self-management in young adults? An integrative review. Nursing Praxis
in Aotearoa New Zealand. 2022;38(2):32-43.
68. Ifediora AC, Obeagu EI, Akahara IC, Eguzouwa UP. Prevalence of urinary tract infection
in diabetic patients attending Umuahia health care facilities. J Bio Innov. 2016;5(1):68-82.
links/5ae45fdfaca272ba507eb3c3/PREVALENCE-OF-URINARY-TRACT-
INFECTION-IN-DIABETIC-PATIENTS-ATTENDING-UMUAHIA-HEALTH-CARE-
FACILITIES.pdf.
69. Ugwu OP, Alum EU, Okon MB, Aja PM, Obeagu EI, Onyeneke EC. Ethanol root extract
and fractions of Sphenocentrum jollyanum abrogate hyperglycaemia and low body weight
in streptozotocin-induced diabetic Wistar albino rats. RPS Pharmacy and Pharmacology
Reports. 2023;2(2):rqad010.
70. Obeagu EI, Obeagu GU. Utilization of Antioxidants in the management of diabetes
mellitus patients. J Diabetes Clin Prac. 2018;1(102):2.
links/5b6c2dec92851ca65053b74e/Utilization-of-Antioxidants-in-the-Management-of-
Diabetes-Mellitus.pdf.
71. Obeagu EI, Okoroiwu IL, Obeagu GU. Some haematological variables in insulin
dependent diabetes mellitus patients in Imo state Nigeria. Int. J. Curr. Res. Chem. Pharm.
Sci. 2016;3(4):110-7. links/5ae4abee458515760ac07a13/Some-haematological-variables-
in-insulin-dependent-diabetes-mellitus-patients-in-Imo-state-Nigeria.pdf.
72. Nwakuilite A, Nwanjo HU, Nwosu DC, Obeagu EI. Evaluation of some trace elements in
streptozocin induced diabetic rats treated with Moringa oleifera leaf powder. WJPMR.
2020;6(12):15-8. links/5fcb587092851c00f8516430/EVALUATION-OF-SOME-

Citation: Obeagu EI, Obeagu GU. Immune Checkpoint Inhibitors in Type 1 Diabetes: A New
Frontier in Therapy. Elite Journal of Medicine, 2024; 2(2): 26-41

13
Elite Journal of Medicine. Volume 2 issue 2(2024), Pp. 26-41
https://epjournals.com/journals/EJM

TRACE-ELEMENTS-IN-STREPTOZOCIN-INDUCED-DIABETIC-RATS-TREATED-
WITH-MORINGA-OLEIFERA-LEAF-POWDER.pdf.
73. Anyiam AF, Obeagu EI, Obi E, Omosigho PO, Irondi EA, Arinze-Anyiam OC, Asiyah
MK. ABO blood groups and gestational diabetes among pregnant women attending
University of Ilorin Teaching Hospital, Kwara State, Nigeria. International Journal of
Research and Reports in Hematology. 2022;5(2):113-121.
74. Okafor CJ, Yusuf SA, Mahmoud SA, Salum SS, Vargas SC, Mathew AE, Obeagu EI,
Shaib HK, Iddi HA, Moh’d MS, Abdulrahman WS. Effect of Gender and Risk Factors in
Complications of Type 2 Diabetic Mellitus among Patients Attending Diabetic Clinic in
Mnazi Mmoja Hospital, Zanzibar. Journal of Pharmaceutical Research International.
2021;33(29B):67-78.
75. Galano ES, Yusuf SA, Ogbonnia SO, Ogundahunsi OA, Obeagu EI, Chukwuani U, Okafor
CJ, Obianagha NF. Effect of Extracts of Kigelia Africana Fruit and Sorghum Bicolor Stalk
on the Biochemical Parameters of Alloxan-Induced Diabetic Rats. Journal of
Pharmaceutical Research International. 2021;33(25B):86-97.
76. Kama SC, Obeagu EI, Alo MN, Ochei KC, Ezugwu UM, Odo M, Ikpeme M, Ukeekwe
CO, Amaeze AA. Incidence of Urinary Tract Infection among Diabetic Patients in
Abakaliki Metropolis. Journal of Pharmaceutical Research International. 2020 Nov
17;32(28):117-121.
77. Nwakulite A, Obeagu EI, Eze R, Vincent CC, Chukwurah EF, Okafor CJ, Ibekwe AM,
Adike CN, Chukwuani U, Ifionu BI. Evaluation of Catalase and Manganese in Type 2
Diabetic Patients in University of Port Harcourt Teaching Hospital. Journal of
Pharmaceutical Research International. 2021:40-45.
78. Nwakulite A, Obeagu EI, Nwanjo HU, Nwosu DC, Nnatuanya IN, Vincent CC, Amaechi
CO, Ochiabu O, Barbara MT, Ibekwe AM, Okafor CJ. Studies on Pancreatic Gene
Expression in Diabetic Rats Treated with Moringa oleifera Leaf. Journal of Pharmaceutical
Research International. 2021;33(28A):78-86.
79. Nwosu DC, Nwanjo HU, Obeagu EI, Ugwu GU, Ofor IB, Okeke A, Ochei KC, Kanu SN,
Okpara KE. Evaluation of Lipoprotein A and Lipid Tetrad Index Pattern in Diabetic
Patients Attending Metabolic Clinic in The Federal Medical Centre, Owerri, Imo State.
World Journal of Pharmacy and Pharmaceutical Sciences, 2015; 4 (3):126-140
80. Ezema GO, Omeh NY, Egbachukwu S, Agbo EC, Ikeyi AP, Obeagu EI. Evaluation of
Biochemical Parameters of Patients with Type 2 Diabetes Mellitus Based on Age and
Gender in Umuahia. Asian Journal of Dental and Health Sciences. 2023 Jun 15;3(2):32-
36. http://ajdhs.com/index.php/journal/article/view/43.
81. Adu ME, Chukwuani U, Ezeoru V, Okafor CJ, Amaechi CO, Vincent CC, Obeagu GU,
Eze R, Nnatuanya IN, Nwosu DC, Nwanjo HU. Studies on molecular docking of moringa
oleifera leaf phytochemical constituents on alpha glucosidase, alpha amylase and
dipeptidyl peptidase. Journal of Pharmaceutical Research International.
2021;33(28A):239-345.
82. Ezugwu UM, Onyenekwe CC, Ukibe NR, Ahaneku JE, Obeagu EI. Plasma Level of
Macromolecules and Mathematical Calculation of Potential Energy in Type 2 Diabetic

Citation: Obeagu EI, Obeagu GU. Immune Checkpoint Inhibitors in Type 1 Diabetes: A New
Frontier in Therapy. Elite Journal of Medicine, 2024; 2(2): 26-41

14
Elite Journal of Medicine. Volume 2 issue 2(2024), Pp. 26-41
https://epjournals.com/journals/EJM

Individuals at NAUTH, Nnewi, Nigeria. Journal of Pharmaceutical Research International.


2021;33(47B):242-248.
83. Nwakulite A, Obeagu EI, Eze R, Ugochi VE, Vincent CC, Okafor CJ, Chukwurah EF,
Unaeze BC, Amaechi CO, Okwuanaso CB, Chukwuani U. Estimation of Serum
Glutathione Peroxidase in Streptozotocin Induced Diabetic Rat Treated with Bitter Leaf
Extract. Journal of Pharmaceutical Research International. 2021;33(30B):200-206.
84. Okoroiwu IL, Obeagu EI, San Miguel HG, Bote SA, Obeagu GU. Characterisation of
HLA-DR antigen in patients type 1 diabetes mellitus in patient attending a tertairy hospital
in Enugu, south-east Nigeria. ACADEMIC JOURNAL. 2023.
85. Okoroiwu IL, Obeagu EI, Obeagu GU, Chikezie CC, Ezema GO. The prevalence of
selected autoimmune diseases. Int. J. Adv. Multidiscip. Res. 2016;3(3):9-14.
86. Nwakuilite A, Nwanjo HU, Nwosu DC, Obeagu EI. EVALUATION OF ENZYME
ANTIOXIDANTS IN STREPTOZOCIN INDUCED DIABETIC RATS TREATED
WITH MORINGA OLEIFERA LEAF POWDER. European Journal of Biomedical.
2020;7(11):285-288.
87. Nwosu DC, Nwanjo HU, Opara AU, Ofor IB, Obeagu EI, Ugwu GU, Ojiegbe GC, Nnorom
RM, Nwokike GI, Okpara KE, Ochei KC. EVALUATION OF C-REACTIVE PROTEIN,
SELENIUM AND GLYCOSYLATED HAEMOGLOBIN LEVELS IN DIABETIC
PATIENTS ATTENDING METABOLIC CLINIC IN THE FEDERAL MEDICAL
CENTRE, OWERRI, IMO STATE. World Journal of Pharmacy and Pharmaceutical
Sciences, 2015; 4 (3):141-152.
https://www.academia.edu/download/38320132/NWOSU_EMMA_9.pdf.
88. Nwakuilite A, Nwanjo HU, Nwosu DC, Obeagu EI. EVALUATION OF KIDNEY
INJURY MOLECULE-1, CYSTATIN C, AND SERUM ELECTROLYTES IN
STREPTOZOCIN INDUCED DIABETIC RATS TREATED WITH MORINGA
OLEIFERA LEAF POWDER. Education. 2002.
89. Ugwu OP, Alum EU, Okon MB, Aja PM, Obeagu EI, Onyeneke EC. Anti-nutritional and
gas chromatography-mass spectrometry (GC-MS) analysis of ethanol root extract and
fractions of Sphenocentrum jollyanum. RPS Pharmacy and Pharmacology Reports.
2023;2(2): rqad007.
90. Obeagu EI, Scott GY, Amekpor F, Ugwu OP, Alum EU. Covid-19 Infection and Diabetes:
A Current Issue. International Journal of Innovative and Applied Research. 2023;11(1):25-
30.
91. Ugwu OP, Alum EU, Obeagu EI, Okon MB, Aja PM, Samson AO, Amusa MO, Adepoju
AO. Effect of Ethanol leaf extract of Chromolaena odorata on lipid profile of streptozotocin
induced diabetic wistar albino rats. IAA Journal of Biological Sciences. 2023;10(1):109-
117.
92. Ifeanyi OE. Gestational Diabetes: Haematological Perspective. South Asian Research
Journal of Applied Medical Sciences, 1 (2):41-42. DOI:
10.36346/SARJAMS.2019.v01i02.003
https://sarpublication.com/media/articles/SARJAMS_12_41-42.pdf.
93. Ogbu IS, Odeh EJ, Ifeanyichukwu OE, Ogbu C, Ude UA, Obeagu EI. Prevalence of
prediabetes among first degree relatives of type 2 diabetes individuals in Abakaliki, Ebonyi
Citation: Obeagu EI, Obeagu GU. Immune Checkpoint Inhibitors in Type 1 Diabetes: A New
Frontier in Therapy. Elite Journal of Medicine, 2024; 2(2): 26-41

15
Elite Journal of Medicine. Volume 2 issue 2(2024), Pp. 26-41
https://epjournals.com/journals/EJM

State Nigeria. Academic Journal of Health Sciences: Medicina Balear. 2023;38(2):85-88.


https://dialnet.unirioja.es/servlet/articulo?codigo=8845439.
94. Ifeanyi OE. An update on Diabetes Mellitus. Int. J. Curr. Res. Med. Sci. 2018;4(6):71-
81.DOI: 10.22192/ijcrms.2018.04.06.012 links/5b3b97a04585150d23f63e76/An-update-
on-Diabetes-Mellitus.pdf.

Citation: Obeagu EI, Obeagu GU. Immune Checkpoint Inhibitors in Type 1 Diabetes: A New
Frontier in Therapy. Elite Journal of Medicine, 2024; 2(2): 26-41

16

View publication stats

You might also like