Print ISSN: 2476-535X, Online ISSN: 2717-2910

Document Type : Review Article

Author

Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore, 641 021, Tamil Nadu, India

Abstract

Marburg virus disease is a rare, but severe illness caused by highly pathogenic Marburg virus, a member of the Filoviridae family. The virus was first identified in 1967 in Marburg and Frankfurt, Germany, as well as Belgrade, Serbia. Since then, sporadic outbreaks have been reported in Central and East Africa. The fruit bats of the genus Rousettus are the major reservoir for the virus. The largest recorded outbreak occurred in Angola in 2005, with 374 cases and 329 deaths, resulting in a mortality rate of approximately 88%, underscoring this virus's potential for causing a devastating impact. Historically, the Marburg virus has received less attention compared to the Ebola virus. However, the geographical expansion of this virus in new regions such as Guinea, Equatorial Guinea, Ghana, and Tanzania highlights its growing threat. The sporadic outbreaks of this deadly pathogen necessitate continued investment in research, surveillance, and public health preparedness to mitigate the impact of this virus on global health security. This review is intended to provide an overview of our current knowledge of the Marburg virus, which is crucial for the development of Marburg virus countermeasures.
 

Keywords

Main Subjects

Introduction

Viral pathogens present a significant burden on global health, causing a range of illnesses from mild infections to severe, high-mortality outbreaks. Emerging and re-emerging viruses continually pose new challenges to global health security (1, 2). Marburg virus (MARV) is one of the highly pathogenic viruses that belong to the order Mononegavirales and is a member of the Filoviridae family (3). It is responsible for severe hemorrhagic fever outbreaks in humans and has emerged as a significant human health threat. MARV was first identified in 1967 during  outbreaks in Marburg and Frankfurt, Germany, as well as Belgrade in Yugoslavia (now Serbia) (4). MARV has since been linked to sporadic outbreaks in Africa, posing significant public health challenges due to its high mortality rate. MARV is primarily endemic to sub-Saharan Africa, particularly Uganda, Angola, and the Democratic Republic of Congo (DRC), where most outbreaks have occurred. The outbreak in DRC (1998-2000) and in Angola (2004 - 2005) demonstrated a mortality rate of 83% and 90%, respectively (5). MARV is listed as the National Institute of Allergy and Infectious Diseases (NIAID) biodefense pathogen (6).

The virus is maintained in wildlife such as bats, particularly species of the Rousettus genus, which serve as reservoir hosts (7). The fruit bats like Rousettus aegyptiacus (Pteropodidae family) are known to carry MARV(8, 9). Infected bats do not typically exhibit symptoms of Marburg virus disease (MVD). This makes them efficient reservoirs as they can spread the virus to other bats or potentially to other animals, including humans, through their saliva, urine, or feces without showing signs of illness (10). These bats are found in parts of Africa, where MARV outbreaks have occurred, such as Uganda, DRC, and Angola. Human infections typically result from contact with infected bats, or through exposure to body fluids of infected individuals during outbreaks (11).

 

Viral Genome

MARV is a single-stranded, enveloped, non-segmented, negative-sense RNA virus that belongs to the genus Marburg virus. Its genome size is about 19 kb in length which encodes for seven structural proteins, including nucleoprotein (NP), virion protein 35 (VP35), VP40, VP30, VP24, glycoprotein (GP), and large (L) viral polymerase (12, 13). Each gene has conserved transcriptional start and stop signals and plays important roles in viral replication and pathogenesis. NP binds to viral RNA, forming the nucleocapsid, while VP35 aids in RNA synthesis and immune evasion. VP40 facilitates viral assembly and budding, whereas GP mediates host cell attachment and entry and is a key target for vaccine and therapeutic development.  VP30 assists in transcription activation, VP24 regulates viral replication, and L functions as the RNA-dependent RNA polymerase for genome replication and transcription (14). During replication, MARV initially attaches to host cells via., GP, enters through endocytosis or membrane fusion, and releases its genomic RNA into the cytoplasm. The polymerase then transcribes mRNA from the negative-sense genome, which is translated into viral proteins by the host cell machinery. The viral genome replication occurs through the production of positive-sense RNA intermediates which acts as a template for generating new viral genomes and mRNA. Viral assembly takes place at the host cell membrane, facilitated by VP40 and GP, leading to budding of mature virions(15, 16).

 

Epidemiology

The recorded outbreaks of MARV have been infrequent yet significantly impactful due to the disease's severity and high mortality rates. MARV outbreaks have been reported mostly in Central and East Africa (Table 1; 11, 17,18). Countries that have experienced outbreaks include Uganda, DRC, Kenya, Angola, Guinea, South Africa and Tanzania. Outbreaks tend to be smaller in scale compared to Ebola virus outbreaks, but can still have significant public health implications due to the severity of illness and high fatality rate (11, 17, 19).

The first recognized outbreak occurred in 1967 in Marburg and Frankfurt, Germany, as well as in Belgrade, Serbia (formerly Yugoslavia), where laboratory workers handling African green monkeys (Cercopithecus aethiops) from Uganda contracted the virus (11). Subsequent outbreaks in 1975 in South Africa, 1980 and 1987 in Kenya, and multiple occurrences in the DRC from 1998 to 2000 underscored the sporadic but constant threat posed by the virus. The largest documented outbreak occurred in Angola in 2005, with 374 reported cases and a mortality rate exceeding 88% (20). This outbreak highlighted significant human-to-human transmission within healthcare settings, emphasizing the critical need for stringent infection control measures. After this outbreak, no other severe fatal outbreaks were documented other than certain sporadic cases. Recurrent outbreaks in Uganda were reported in 2008-2009, 2012, 2014, and 2017. The first reported outbreak of MVD in Tanzania occurred in 2022, involving a total of eight cases, with five fatalities (11, 17).

 

 

Transmission and Symptoms

The available evidence to date suggests that the virus is transmitted to humans via, direct or indirect contact with Rousettus bats (8). Direct exposure to infected bats or their bodily fluids is a primary route of transmission (21). Subsequent human-to-human transmission occurs through direct contact with the infected person’s bodily fluids such as blood, saliva, vomit, urine, feces, respiratory secretions, and semen (11).

MVD manifests with an incubation period ranging from 2-21 days (22), followed by abrupt onset of fever, headache, myalgia, and gastrointestinal symptoms. The illness typically begins with a high fever (> 39°C) accompanied by intense chills, headache, and profound malaise (23). Muscle and joint pain contribute to the discomfort of the affected individuals. Gastrointestinal symptoms such as nausea, vomiting, and diarrhea can quickly lead to dehydration, exacerbating the severity of the disease. Respiratory symptoms may include chest pain, cough, and sore throat. In more severe cases, patients may develop hemorrhagic manifestations, though bleeding is less frequent compared to other viral hemorrhagic fevers.  Between 5 to 7 days after infection, many patients experience severe hemorrhagic symptoms (24). Neurological symptoms, such as confusion, agitation, and seizures, may occur in advanced stages. Severe cases progress to multi-organ dysfunction, hemorrhagic manifestations, and shock, with a case fatality rate ranging from 24% to 88% (25). Early detection and isolation of cases are essential to prevent further spread of the virus in affected communities (25).

 

Diagnosis and Treatment

Given the potential for rapid disease progression and severe outcomes associated with MARV infection, early diagnosis is critical for initiating appropriate patient management and implementing infection control measures. The diagnosis of MVD requires laboratory testing due to the similarity of its early symptoms to those of other infectious diseases, such as malaria, typhoid, and other viral hemorrhagic fevers(26).  MARV infection diagnosis relies on detecting viral RNA in blood or tissues using reverse transcription polymerase chain reaction (RT-PCR;27). Serological tests, including enzyme-linked immunosorbent assays (ELISA), detect antibodies against MARV antigens, serum neutralization test, and virus isolation by cell culture (28-30). The samples should be tested in maximum biological containment conditions. The rapid diagnostic tests are essential for early disease identification and outbreak control in resource-limited settings.

There are no specific antiviral therapies or vaccines approved presently for MARV, leaving supportive care as the primary treatment option. Fluid and electrolyte replacement, symptomatic relief, and intensive care support may improve the condition significantly (31). Advances in supportive care have improved the survival outcomes of the affected individuals. The development of effective vaccines and treatment options is highly essential to control the virus (11).

 

Prevention and Control Strategies

The prevention of MARV outbreaks relies on proactive measures at various levels, from individual protective behaviors to national and international public health interventions. The virus has the potential to spread globally due to international travel and trade, necessitating robust surveillance and response capabilities to detect and contain outbreaks promptly.  As the world is slowly recovering from the COVID-19 pandemic, there are now concerning reports indicating the increasing cases of monkeypox and SARS-CoV-2 variants across multiple countries (32, 33). The international community must remain vigilant regarding MARV, a highly contagious and lethal pathogen. If this virus spreads globally, it will have a devastating impact on the human population. Hence, effective preparedness response efforts and community engagement are crucial to mitigate the public health impact of MARV outbreaks. This includes strengthening the surveillance systems to detect early signs of outbreaks, enhancing laboratory capacity for rapid diagnosis, establishing emergency response plans, training healthcare workers in prevention, and control measures, and accelerating research into vaccines and treatments. Public awareness campaigns and community engagement are also essential to promote understanding of the virus, encourage timely healthcare-seeking behavior, and foster cooperation with outbreak response activities (34).

It is important to follow stringent infection control practices in healthcare settings, including the use of personal protective equipment (PPE), hand hygiene, and isolation protocols for confirmed/susceptible cases. The collected samples in testing laboratories should be managed by trained personnel and processed carefully in well-equipped laboratories. Additionally, ensuring proper use, disinfection, and appropriate disposal of instruments and equipment used in patient care are essential components of comprehensive infection control measures (11). Several studies are ongoing to develop vaccines against MARV. Ongoing vaccine studies and trials for MARV utilize various platforms to induce protective immune responses (35). Vaccines targeting MARV glycoprotein are in development, showing promise in preclinical studies and phase 1 clinical trials (36). The recombinant vesicular stomatitis virus (rVSV), adenovirus vector vaccines, DNA vaccines,  virus-like particle (VLP) vaccines, mRNA vaccines, and protein subunit vaccines are also being explored for their potential to protect against MARV (36-44). Continued research and clinical trials are essential to advance the promising vaccine candidates toward licensure and deployment to protect at-risk populations.

Conclusion

Emerging infectious diseases significantly impact public health and the global economy. MARV represents a significant public health concern due to its high mortality rate, potential for global spread, limited treatment options, and impact on vulnerable healthcare systems and communities. Due to the increase in the incidence of MARV infections, there is an immediate need to develop effective diagnostic tools, therapeutics, and vaccines for improving outcomes and reducing the burden of MVD in endemic regions. Considering the intensity of past outbreaks, MARV has the potential to cause severe outbreaks, if not properly controlled. The epidemiologists have identified MARV as a significant threat to global public health. Therefore, increased research and focus on this virus are imperative for mitigating potential future outbreaks. Hence, a concerted global effort is needed to strengthen surveillance, enhance healthcare infrastructure, and promote interdisciplinary collaboration to effectively combat this deadly pathogen.

 

Acknowledgments

The author is very thankful to the Karpagam Academy of Higher Education for their support.

Ethical approval

Not applicable.

Conflicts of Interest

The author declares no conflict of interest.

  1. References

    1. Zumla A, Hui DSC. Emerging and reemerging infectious diseases: Global overview. Infect Dis Clin North Am. 2019;33(4):xiii-xix. doi:10.1016/j.idc.2019.09.001.
    2. Shanmugaraj B, Kothalam R, Mohamed Sheik TAA. A brief overview of the threat of zoonotic viruses. Microbes and Infectious Diseases. 2024: doi: 10.21608/mid.2024.294905.1975.
    3. Kuhn JH, Amarasinghe GK, Basler CF, Bavari S, Bukreyev A, Chandran K, et al. Virus taxonomy profile: Filoviridae. J Gen Virol. 2019;100(6):911-912. doi:10.1099/jgv.0.001252.
    4. Luby JP, Sanders CV. Green monkey disease ("Marburg virus" disease): a new zoonosis. Ann Intern Med. 1969;71(3):657-60.
    5. Languon S, Quaye O. Filovirus disease outbreaks: A chronological overview. 2019;10:1178122x19849927. doi:10.1177/1178122X19849927.
    6. NIAID biodefense pathogens 2024 [cited 2024 June 25]. Available from: https://www.niaid.nih.gov/research/niaid-biodefense-pathogens#k.
    7. Makenov MT, Boumbaly S, Tolno FR, Sacko N, N'Fatoma LT, Mansare O, et al. Marburg virus in Egyptian Rousettus bats in Guinea: Investigation of Marburg virus outbreak origin in 2021. PLoS Negl Trop Dis. 2023;17(4):e0011279. doi:10.1371/journal.pntd.0011279.
    8. Towner JS, Amman BR, Sealy TK, Carroll SA, Comer JA, Kemp A, et al. Isolation of genetically diverse Marburg viruses from Egyptian fruit bats. PLoS Pathog. 2009;5(7):e1000536. doi:10.1371/journal.ppat.1000536
    9. Amman BR, Jones ME, Sealy TK, Uebelhoer LS, Schuh AJ, Bird BH, et al. Oral shedding of Marburg virus in experimentally infected Egyptian fruit bats (Rousettus aegyptiacus). J Wildl Dis. 2015;51(1):113-24. doi:10.7589/2014-08-198.
    10. Guito JC, Kirejczyk SGM, Schuh AJ, Amman BR, Sealy TK, Graziano J, et al. Coordinated inflammatory responses dictate Marburg virus control by reservoir bats. Nat Commun. 2024;15(1):1826. doi:https://doi.org/10.1038/s41467-024-46226-7.
    11. Marburg virus disease 2021 [cited 2024 June 25]. Available from: https://www.who.int/news-room/fact-sheets/detail/marburg-virus-disease.
    12. Feldmann H, Kiley MP. Classification, structure, and replication of filoviruses. Curr Top Microbiol Immunol. 1999;235:1-21. doi:10.1007/978-3-642-59949-1_1.
    13. Mühlberger E. Filovirus replication and transcription. Future Virol. 2007;2(2):205-15. doi:10.2217/17460794.2.2.205.
    14. Feldmann H, Mühlberger E, Randolf A, Will C, Kiley MP, Sanchez A, et al. Marburg virus, a filovirus: messenger RNAs, gene order, and regulatory elements of the replication cycle. Virus Res. 1992;24(1):1-19. doi: https://doi.org/10.1016/0168-1702(92)90027-7.
    15. Abir MH, Rahman T, Das A, Etu SN, Nafiz IH, Rakib A, et al. Pathogenicity and virulence of Marburg virus. Virulence.2022;13(1):609-633. doi:10.1080/21505594.2022.2054760.
    16. Schmidt KM, Mühlberger E. Marburg virus reverse genetics systems. Viruses. 2016;8(6):178. doi:10.3390/v8060178
    17. History of Marburg Disease Outbreaks 2024 [cited 2024 June 25]. Available from: https://www.cdc.gov/marburg/outbreaks/index.html.
    18. Srivastava D, Kutikuppala LVS, Shanker P, Sahoo RN, Pattnaik G, Dash R, et al. The neglected continuously emerging Marburg virus disease in Africa: A global public health threat. Health Sci Rep.2023;6(11):e1661. doi:10.1002/hsr2.1661.
    19. Brauburger K, Hume AJ, Mühlberger E, Olejnik J. Forty-five years of Marburg virus research. Viruses. 2012;4(10):1878-927. doi:10.3390/v4101878.
    20. Outbreak of Marburg virus hemorrhagic fever--Angola, October 1, 2004-March 29, 2005. MMWR Morbidity and mortality weekly report. 2005;54(12):308-9.
    21. Nyakarahuka L, Shoemaker TR, Balinandi S, Chemos G, Kwesiga B, Mulei S, et al. Marburg virus disease outbreak in Kween District Uganda, 2017: Epidemiological and laboratory findings. PLoS Negl Trop Dis. 2019;13(3):e0007257. doi:10.1371/journal.pntd.0007257.
    22. Deb N, Roy P, Jaiswal V, Mohanty A, Sah S, Sah R. Marburg Virus Disease in Tanzania: The most recent outbreak. New Microbes New Infect. 2023;53:101123. doi:10.1016/j.nmni.2023.101123.
    23. Gear JS, Cassel GA, Gear AJ, Trappler B, Clausen L, Meyers AM, et al. Outbreake of Marburg virus disease in Johannesburg. Br Med J. 1975;4(5995):489-93. doi:10.1136/bmj.4.5995.489.
    24. Cobo F. Viruses causing hemorrhagic fever. Safety laboratory procedures. Open Virol J. 2016;10:1-9. doi:10.2174/1874357901610010001.
    25. Srivastava S, Sharma D, Kumar S, Sharma A, Rijal R, Asija A, et al. Emergence of Marburg virus: a global perspective on fatal outbreaks and clinical challenges. Front Microbiol. 2023;14 :1239079. doi:10.3389/fmicb.2023.1239079.
    26. Flórez-Álvarez L, de Souza EE, Botosso VF, de Oliveira DBL, Ho PL, Taborda CP, et al. Hemorrhagic fever viruses: Pathogenesis, therapeutics, and emerging and re-emerging potential. Front Microbiol. 2022;13:1040093. doi:10.3389/fmicb.2022.1040093.
    27. Drosten C, Göttig S, Schilling S, Asper M, Panning M, Schmitz H, et al. Rapid detection and quantification of RNA of Ebola and Marburg viruses, Lassa virus, Crimean-Congo hemorrhagic fever virus, Rift Valley fever virus, dengue virus, and yellow fever virus by real-time reverse transcription-PCR. J Clin Microbiol. 2002;40(7):2323-2330. doi:10.1128/JCM.40.7.2323-2330.2002.
    28. Ksiazek TG, Rollin PE, Jahrling PB, Johnson E, Dalgard DW, Peters CJ. Enzyme immunosorbent assay for Ebola virus antigens in tissues of infected primates. J Clin Microbiol. 1992;30(4):947-50. doi:10.1128/jcm.30.4.947-950.1992.
    29. Ksiazek TG, West CP, Rollin PE, Jahrling PB, Peters CJ. ELISA for the detection of antibodies to Ebola viruses. The Journal of infectious diseases. 1999;179 Suppl 1:S192-S198. doi:10.1086/514313.
    30. Grolla A, Lucht A, Dick D, Strong JE, Feldmann H. Laboratory diagnosis of Ebola and Marburg hemorrhagic fever. Bull Soc Pathol Exot. 2005;98(3):205-209. PMID: 16267962.
    31. Factsheet about Marburg virus disease 2024 [cited 2024 June 25]. Available from:https://www.ecdc.europa.eu/en/infectious-disease-topics/ebola-virus-disease/facts/factsheet-about-marburg-virus-disease.
    32. Cheema SA, Munir T, Ullah K, Kifayat T, Rahman A, Emam W, et al. Trends in Monkeypox transmission: Investigation into 30 most affected countries. Heliyon. 2024;10(1):e21980. doi: 10.1016/j.heliyon.2023.e21980.
    33. Shanmugaraj B. Ever-evolving SARS-CoV-2: Latest variant KP.2 is on the rise. Asian Pac J Trop Med. 2024;17(6). doi: 10.4103/apjtm.apjtm_341_24
    34. Aderinto N. A reflection on the Marburg virus outbreak in Tanzania: the importance of preparedness and prevention in public health - a correspondence. Ann Med Surg (Lond). 2023;85(5):2247-2249. doi:10.1097/MS9.0000000000000596.
    35. Kortepeter MG, Dierberg K, Shenoy ES, Cieslak TJ. Marburg virus disease: A summary for clinicians. Int J Infect Dis. 2020;99:233-242. doi:10.1016/j.ijid.2020.07.042.
    36. Hamer MJ, Houser KV, Hofstetter AR, Ortega-Villa AM, Lee C, Preston A, et al. Safety, tolerability, and immunogenicity of the chimpanzee adenovirus type 3-vectored Marburg virus (cAd3-Marburg) vaccine in healthy adults in the USA: a first-in-human, phase 1, open-label, dose-escalation trial. Lancet. 2023;401(10373):294-302. doi: https://doi.org/10.1016/S0140-6736(22)02400-X.
    37. Lehrer AT, Chuang E, Namekar M, Williams CA, Wong TAS, Lieberman MM, et al. Recombinant protein filovirus vaccines protect Cynomolgus Macaques from Ebola, Sudan, and Marburg Viruses. Front Immunol. 2021;12:703986. doi:10.3389/fimmu.2021.703986.
    38. Swenson DL, Warfield KL, Larsen T, Alves DA, Coberley SS, Bavari S. Monovalent virus-like particle vaccine protects guinea pigs and nonhuman primates against infection with multiple Marburg viruses. Expert Rev Vaccines. 2008;7(4):417-429. doi:10.1586/14760584.7.4.417
    39. Milligan ID, Gibani MM, Sewell R, Clutterbuck EA, Campbell D, Plested E, et al. Safety and Immunogenicity of novel adenovirus type 26- and modified vaccinia ankara-vectored Ebola vaccines: A randomized clinical trial. JAMA. 2016;315(15):1610-1623. doi:10.1001/jama.2016.4218.
    40. Kibuuka H, Berkowitz NM, Millard M, Enama ME, Tindikahwa A, Sekiziyivu AB, et al. Safety and immunogenicity of Ebola virus and Marburg virus glycoprotein DNA vaccines assessed separately and concomitantly in healthy Ugandan adults: a phase 1b, randomised, double-blind, placebo-controlled clinical trial. Lancet (London, England). 2015;385(9977):1545-1554. doi:https://doi.org/10.1016/S0140-6736(14)62385-0.
    41. Sarwar UN, Costner P, Enama ME, Berkowitz N, Hu Z, Hendel CS, et al. Safety and immunogenicity of DNA vaccines encoding Ebolavirus and Marburgvirus wild-type glycoproteins in a phase I clinical trial. J Infect Dis. 2015;211(4):549-557. doi:10.1093/infdis/jiu511.
    42. Cooper CL, Morrow G. Nonhuman primates are protected against marburg virus disease by vaccination with a vesicular stomatitis virus vector-based vaccine prepared under conditions to allow advancement to human clinical trials. Vaccines (Basel). 2022;10(10) :1582. doi:10.3390/vaccines10101582.
    43. Geisbert TW, Daddario-Dicaprio KM, Geisbert JB, Reed DS, Feldmann F, Grolla A, et al. Vesicular stomatitis virus-based vaccines protect nonhuman primates against aerosol challenge with Ebola and Marburg viruses. Vaccine. 2008;26(52):6894-6900. doi:10.1016/j.vaccine.2008.09.082
    44. Zhu W, Liu G, Cao W, He S, Leung A, Ströher U, et al. A cloned recombinant vesicular stomatitis virus-vectored Marburg vaccine, PHV01, protects guinea pigs from lethal marburg virus disease. Vaccines (Basel).2022;10(7):1004. doi:10.3390/vaccines10071004.