Skip to main content
Clinical Microbiology Reviews logoLink to Clinical Microbiology Reviews
. 2022 Jun 21;35(3):e00211-21. doi: 10.1128/cmr.00211-21

Diagnosis, Management, and Future Control of Cholera

Fahima Chowdhury a,b,, Allen G Ross c, Md Taufiqul Islam a,b, Nigel A J McMillan b, Firdausi Qadri a
PMCID: PMC9491185  PMID: 35726607

SUMMARY

Cholera, caused by Vibrio cholerae, persists in developing countries due to inadequate access to safe water, sanitation, and hygiene. There are approximately 4 million cases and 143,000 deaths each year due to cholera. The disease is transmitted fecally-orally via contaminated food or water. Severe dehydrating cholera can progress to hypovolemic shock due to the rapid loss of fluids and electrolytes, which requires a rapid infusion of intravenous (i.v.) fluids. The case fatality rate exceeds 50% without proper clinical management but can be less than 1% with prompt rehydration and antibiotics. Oral cholera vaccines (OCVs) serve as a major component of an integrated control package during outbreaks or within zones of endemicity. Water, sanitation, and hygiene (WaSH); health education; and prophylactic antibiotic treatment are additional components of the prevention and control of cholera. The World Health Organization (WHO) and the Global Task Force for Cholera Control (GTFCC) have set an ambitious goal of eliminating cholera by 2030 in high-risk areas.

KEYWORDS: Vibrio cholerae, WaSH, oral cholera vaccine, treatment

INTRODUCTION

Vibrio cholerae is a major cause of severe dehydrating diarrheal disease and remains a major public health problem in low- and middle-income countries (LMICs) where water, sanitation, and hygiene (WaSH) are inadequate (1). Severe cholera can rapidly lead to hypovolemic shock after the onset of diarrhea and vomiting. However, with appropriate oral and intravenous (i.v.) fluid therapy, clinicians can lower the case fatality rate from over 50% to less than 1% (2). There are two fecal-oral mechanisms of V. cholerae transmission: one involves direct spread from person to person as a consequence of eating bacterium-contaminated food or water, and the other involves drinking environmentally polluted water from ponds, lakes, or rivers (3, 4). Fomites and flies may help disseminate V. cholerae by acting as mechanical vectors (3).

The World Health Organization (WHO) approximates 1.4 million to 4.0 million cases and 21,000 to 143,000 deaths globally due to the disease per annum (5). A zone where cholera is endemic is defined as an area with confirmed cases detected over the past 3 years (5). Cholera is endemic in Asia, Latin and Central America, and sub-Saharan Africa (1, 5, 6). V. cholerae originated in the Ganges River Delta and remains highly prevalent in Asia and Africa (7). Outbreaks of cholera are unpredictable and can occur in both zones where cholera is endemic and those where it is nonendemic depending on environmental conditions, with refugee camps being particularly susceptible (8). V. cholerae is always evolving, with new phenotypes and genotypes emerging with outbreaks and as a result of increased antibiotic resistance (9). The WHO’s Global Task Force for Cholera Control (GTFCC) recommends access to both oral cholera vaccination and improved WaSH to avert cholera transmission as well as continued diarrheal disease surveillance (10).

EPIDEMIOLOGY

Geographical Distribution and Burden of Cholera

In resource-poor countries of endemicity, data from the last decade have shown that the burden of cholera has increased, and it has become an important public health problem (8). Figure 1 illustrates the global geographical distribution of cholera between 2016 and 2019 (1114). Cholera has been historically endemic in the Asian subcontinent (e.g., Indonesia, India, Bangladesh, Vietnam, Thailand, Pakistan, Nepal, and Iraq) but is now endemic in Africa (e.g., South Africa, Mozambique, Botswana, Zambia, Sierra Leone, Nigeria, Angola, the Democratic Republic of the Congo [DRC], Yemen, Zimbabwe, the United Republic of Tanzania, and Guinea), Latin America (Brazil, Peru, Chile, Columbia, and Ecuador), and the Caribbean (Haiti, Cuba, and the Dominican Republic) (1114). Due to weak or absent surveillance systems, many countries do not report cholera cases or deaths (3). Some countries are cautious in announcing cholera outbreaks to avoid economic losses in tourism and exports and to prevent general panic in society. But early reporting of cholera outbreaks has resulted in shorter durations of epidemics (15). Since 2006, 52 developing countries have reported increasing numbers of cholera cases (6).

FIG 1.

FIG 1

Global distribution of cholera between 2016 and 2019. ★, the imported cases were from elsewhere in the country where cholera is not common. The red zones represent the affected countries where cholera is endemic during the 3-year period.

In the Bengal Delta region, cholera epidemics typically follow a seasonal pattern, with one peak in spring (March to May) and another following the rainy season (September to November) (1620). In Africa, epidemics occur in different regions during the rainy season, with recent outbreaks being reported in Zanzibar, the Eastern DRC, Angola, and West Africa. Haiti experienced a recent outbreak from 2017 to 2018 due to Hurricane Matthew, and the WHO reported 800,000 cholera cases and approximately 10,000 deaths from cholera since the outbreak (3). Mathematical modeling of cholera transmission suggests that outbreaks are reliant on variations in the environment and herd protection (21).

Risk Factors

The risk factors for acquiring cholera are associated with poverty, including inadequate sanitation, contaminated drinking water, and poor food hygiene (e.g., street foods). Handwashing with soap before and after meals and after defecation is associated with a reduced risk (22).

There are some biological factors that have been recognized as risk factors for cholera, and these are female gender, blood group O, and retinol deficiency, and hypochlorhydria (i.e., in people who take histamine receptor blockers, antacids, and proton pump inhibitors) also increases the risk of contracting cholera (2328). Moreover, Helicobacter pylori infection and gastrectomy are two major factors contributing to severe disease (1). Malnutrition increases susceptibility, especially among young children (1). The incidence of cholera in countries of endemicity is highest among children less than 5 years of age as they have low levels of acquired immunity compared to adults (29).

Secretory immunoglobulin A (SIgA), which is secreted in breast milk, protects against severe cholera, and thus, exclusive breastfeeding for the first 6 months of life is recommended for women living in communities of endemicity (30). Long palate, lung, and nasal epithelium clone protein 1 (LPLUNC1) is expressed in Paneth cells of the intestinal mucosa and is strongly associated with modulating host inflammatory responses to V. cholerae infection and disease severity (25). Concomitant infection of the gut with other bacteria like enterotoxigenic Escherichia coli (ETEC) or parasites increases the chance of V. cholerae infection (31, 32).

A household contact study of cholera patients in Bangladesh showed that first-degree relatives (parents, offspring, and siblings) have a greater chance of acquiring cholera than second-degree relatives (grandchildren, grandparents, uncles, and aunts) staying in the same household (15).

PATHOLOGY

Microbiology and Pathogenesis

V. cholerae is a Gram-negative, comma-shaped bacterium that is classified serologically into over 200 serogroups. Of them, V. cholerae O1 and O139 have caused recent cholera epidemics (33). Based on biochemical structure, V. cholerae O1 is categorized into two biotypes, classical and El Tor, and, more recently, the altered El Tor biotype. Furthermore, each biotype is differentiated into three serotypes, Ogawa, Inaba, and the rare Hikojima type (34).

V. cholerae releases a heat-labile exotoxin based on the AB5 multimeric protein, named cholera toxin (CT), which adheres to the small intestinal mucosa of the gut. CT consists of two subunits (one A subunit [CTA] and five B subunits [CTB]), which cause fluid and electrolyte loss. The B subunit fixes onto eukaryotic cells, whereas the A subunit is shifted into the cell, which assists in increasing cyclic AMP (cAMP) and subsequently leads to secretory diarrhea, which causes severe dehydration. The toxin-coregulated pilus (TcpA), which is required for colonization, serves as a receptor for the cholera toxin phage (CTXphi), which is the V. cholerae-specific filamentous bacteriophage and carries the gene for CT (35).

V. cholerae exists in stagnant water or marine environmental reservoirs where the water source is infected with human and/or animal waste. V. cholerae in freshly shed stool appears to be hyperinfectious for 24 h after release into the environment (36). V. cholerae flourishes in 30°C water with 15% salinity and a pH of 8.5 (37). During spring and after the monsoon season, more adverse environmental conditions can trigger an outbreak of V. cholerae. Real-time recording of climatic parameters along with active surveillance systems can assist public health warning systems in minimizing risk factors (37). The intake of polluted water and contaminated food are the main sources of infection, although acidic gastric enzymes kill most of the bacteria, and the rest colonize the small intestine. The infective dose of V. cholerae is 103 to 108 CFU when ingested with water, but a minimum dose of ~102 to 104 CFU can cause diarrhea when ingested with food. (https://www.medscape.com/answers/962643-54707/what-is-the-infectious-dose-of-vibrio-v-cholerae-required-to-cause-cholera). A high infectious dose, 108 CFU, is required to cause diarrhea in healthy individuals, while a very small inoculum of 105 CFU can cause diarrhea in individuals with low levels of gastric acid (38). The incubation period of V. cholerae may range from 12 h to 5 days (39).

Molecular Epidemiology of V. cholerae

One focus of molecular epidemiological analysis of V. cholerae is the CTX phage, which contains the CT genes. The cholera toxin-encoding genes ctxAB carried by toxigenic V. cholerae have undergone numerous genetic mutations, which include two main components of CTX phage (CTXcla and CTX-1) and repeat sequence 1 (RS1), along with point mutations in ctxB (40). Since 1817, a total of seven cholera pandemics have occurred worldwide, with the most recent pandemic continuing until the present (3). It appears that the O1 classical biotype was the causative agent of the first five pandemics (1817 to 1896). Following this, the O1 classical biotype CTXcla was responsible for the sixth pandemic (1899 to 1923) (40, 41). During the pre-seventh-pandemic period (1923 to 1961), only a few sporadic outbreaks were reported with the El Tor biotype (41). The current cholera pandemic (7th cholera pandemic [7CP]) began in 1961 in Indonesia, with the El Tor biotype as the causative agent, and spread to South Asia after 2 years and then to Africa (1970), South America (1990), and the Caribbean (2010).

A new V. cholerae serogroup, O139, emerged in the Indian subcontinent in 1992 and transmitted throughout the Asian subcontinent by the mid-2000s. In 2004, another new V. cholerae O1 type was isolated in Asia and Africa as a hybrid El Tor biotype encoded with classical CT (40, 41).

Eight diverse phyletic lineages (L1 to L8) have been classified by genomic analysis, based on single nucleotide polymorphisms (SNPs) from the O1 and O139 serogroups. The classical (L1) and El Tor (L2) biotypes were separated into two distinctly evolved lineages. The L1 and L3 to -6 lineages represent the first six pandemics, while the L2 lineage, also known as 7 P V. cholerae El Tor (7PET), is responsible for 7CP having three waves and several phylogenetic sublineages of transmission events (T1 to -13 and Latin American transmission 1 [LAT-1] to LAT-3). T1 to T12 originated from Africa, while T13 originated from East Africa and Yemen (40, 41).

The wave 1 (T1 to -6 and LAT-1 and -2) isolates were composed of CTX-1, the repressor gene (rstR) of the El Tor biotype (rstREl Tor) on chromosome 1, and CT genotype 3 (ctxB3) of V. cholerae O1 added with the toxin-linked cryptic (TLC) component and persisted from 1961 to 1999. Wave 1 spread from Indonesia to Southeast Asia, Mozambique, Angola, the Middle East, East Europe, Ethiopia, Angola, the U.S. Gulf Coast, and Latin America. Wave 2 (T7 and -8) isolates were composed of CT genotype 1 (ctxB1), a recurrence of ctx-2 on chromosome 2, ctx-1, and RS1 on chromosome 1. The wave was prominent between 1978 and 1984. It originated in India and spread to East Asia and Africa. V. cholerae O139 contains ctxB4 to ctxB6 and was prevalent from 1999 to 2005 in Bangladesh. Wave 3 (T9 to -13 and LAT-3) isolates are composed of rstREl Tor and TLC:RS1:CTX3 to TLC:RS1:CTX6 in CT genotype 1 (1991 to 2010) and carry the integrating and conjugative element (ICE) (specifically, ICEVchInd5) of the SXT/R391 type (encoding resistance to chloramphenicol, streptomycin, tetracycline, sulfamethoxazole, and trimethoprim). Wave 3 isolates are divided into three or more types. Wave 3 originated in India in 2006 and caused outbreaks in Haiti, Yemen, and Mariupol, Ukraine, in 2019 (40).

Clinical and Metabolic Manifestations

Cholera infection may be asymptomatic, mild, moderate, or severe (42). Diarrhea in cholera patients is usually painless and may contain bile or fecal matter in the early stages of infection. “Rice water stools” are unique to cholera patients. They are starchy in color, look more like water that contains uncooked rice or has been used to wash rice, and have a fishy odor (42, 43). An adult cholera patient can produce up to 1,000 mL per h of loose watery stool, leading to hypovolemia, shock, and death, termed “cholera gravis.” The rate of excretion rate stool in children with severe cholera is generally between 10 and 20 mL/kg/h (44). This stool contains potassium, sodium, and bicarbonate. Signs of dehydration (e.g., sunken eyes, tears, dry mouth, thirst, rapid pulse, lethargy, cold skin, loss of skin elasticity, or crumpled hands and feet) are present due to profuse watery diarrhea (Table 1). Deep and rapid respiration due to hyperventilation (Kussmaul breathing) and acidosis (loss of bicarbonate in the stool) are some of the striking features of severe cholera. Symptomatic cholera patients can shed bacteria in their stool for 2 days to 2 weeks from the onset of infection (Fig. 2), whereas asymptomatic carriers shed for only a few days (<7 days) (15). In settings where cholera is endemic, spatiotemporal analysis has shown that index cholera cases can be very infectious during the first 5 days of infection and can spread the bacteria within a 200-m radius of their home. Household contacts have a 100-times-higher risk of acquiring cholera than those outside the radius (45).

TABLE 1.

Clinical assessment of dehydrationd

Condition Assessment of dehydration severity
No signs of dehydration Some dehydration Severe dehydration
Physical appearance Well, alert Restless/irritablea Lethargic/unconsciousa
Eyes Normal Sunken Very sunken and dry
Tearsb Present Absent Absent
Mouth and tongue Moist Dry Very dry
Thirst Drinks normally, not thirsty Thirsty, drinks eagerlya Drinks poorly/unable to drinka
Skin pinchc Goes back quickly Goes back slowlya Goes back very slowlya
Radial pulse Normal Rapid, low vola Weak or absenta
Diagnosis No signs of dehydration If the patient has 2 or more signs, including at least 1 signa of mandatory criteria, there is some dehydration If the patient has 2 or more signs, including at least 1 signa of mandatory criteria, there is severe dehydration
a

Mandatory criteria for the diagnosis of different grades of dehydration.

b

Tear sign is applicable only to infants and younger children.

c

The skin pinch is less useful in severely malnourished children or overweight patients.

d

Adapted from WHO guidelines on the management of patients with cholera (58, 144).

FIG 2.

FIG 2

Life cycle of V. cholerae.

Approximately 5 to 10% of patients develop severe dehydration from an increased frequency of profuse watery stool and excessive vomiting, which can rapidly deplete a large volume of water from the body, leading to kidney failure, shock, sepsis, and even death within a few hours if left untreated (1). Electrolyte imbalance is a common complication of cholera, which includes hyponatremia or hypernatremia, hypocalcemia, and hypokalemia (42). Renal failure due to decreased urinary output and aspiration pneumonia are also common in children (46). The accumulation of fluid in the intestinal lumen (cholera sicca) is very uncommon. Children under 5 years of age may develop chronic enteropathy and malnutrition. Inadequate rehydration may cause metabolic abnormalities among patients suffering from severe dehydration (42). Reduced food intake during acute illness may lead to hypoglycemia, a lethal complication that is more common in children (1).

DIAGNOSIS

The diagnosis of cholera is frequently based on clinical signs and symptoms (Table 1) in resource-limited areas of endemicity where laboratory facilities are not available (42). In nonendemic settings, cholera is suspected if a patient has severe dehydration or someone has died from acute watery diarrhea (AWD). But in a cholera epidemic setting, if a patient (>5 years of age) has AWD more than three times with or without vomiting within 24 h, cholera is indicated (47).

Generally, in the developed laboratory setting, for the diagnosis of cholera, stool or rectal swab culture is the gold-standard reference method and costs approximately $10 (40). The specimens are placed into an enrichment broth made of alkaline peptone water, which enhances the sensitivity of the culture, and are later subcultured on selective thiosulfate citrate bile salt (TCBS) agar or taurocholate tellurite gelatin agar (TTGA), which is the ideal culture medium (48). Hence, performing stool culture requires trained personnel and a laboratory facility (49). Cary-Blair medium is commonly used as the medium for transport from field settings to the laboratory (40).

Different methods (e.g., biochemical, immunochemical, or molecular) with specific antiserum or monoclonal antibodies are used to characterize the biochemical properties of V. cholerae O1 such as classical, El Tor, or altered variants (43). Dark-field microscopy can be used to rapidly detect V. cholerae in stool samples before culture (50). However, more than half of the dark-field-microscopy-negative samples are found to be positive for V. cholerae when cultured (51). V. cholerae diagnosis using PCR is highly sensitive, but this technique needs an enhanced laboratory capacity, which is often lacking in most LMICs (43, 46). PCR can be used to detect molecular markers of certain phenotypes with target genes such as ctxA, tcpA, and ompW (52). Even though PCR costs are approximately the same as those of stool culture and PCR requires a specific laboratory setup, the results can be obtained much earlier than with stool culture.

In rural or underdeveloped health care settings where there is a scarcity of culture medium/PCR and/or trained personnel (5), rapid diagnostic tests (RDTs) of stool samples cost only $2, and these can be performed without special training. RDTs can also provide an early warning for public health experts when a cholera outbreak is imminent. Different categories of RDTs are available on the market, with a wide range of sensitivities and specificities. Monoclonal antibodies in Crystal VC (Span Diagnostics Ltd., Surat, India) can easily detect the lipopolysaccharide (LPS) antigens of both V. cholerae O1 and O139 serogroups. Crystal VC has shown 97% sensitivity and 76% specificity (53). Cholkit is available in Bangladesh, with acceptable sensitivity (76%) and good specificity (90%), and is less expensive (less than $2) than the existing RDTs (54). However, this kit detects only the V. cholerae O1 serogroup and hence needs to be used in settings where cholera is endemic and where this serogroup is predominant. Currently, V. cholerae O1 is the predominant serogroup, although V. cholerae O139 strains are occasionally isolated in Bangladesh (3, 55).

CLINICAL MANAGEMENT

Fluid Replacement

Early diagnosis and rapid management of dehydration are crucial for increasing positive outcomes. Most cholera cases are usually mild to moderate and easily managed with an oral rehydration solution (ORS) (https://www.who.int/news-room/fact-sheets/detail/cholera). Currently, the WHO and UNICEF recommend low-osmolarity ORS, which contains sodium, chloride, potassium, citrate, and anhydrous glucose prepared in 1,000 mL of sterile water (Table 2). This improved ORS recipe is safe, lowers hypertonicity, and decreases stool output (56). ORS can also be prepared at home by mixing 1/2 teaspoon of salt and 6 teaspoons of sugar in 1,000 mL of sterile water. Rice-based saline (i.e., rice powder) is also used for those above 6 months of age but is more difficult to prepare (57). After each purging of watery stool, ORS is given according to different age groups to counterbalance the amount of stool loss (Fig. 3). For young children up to 2 years of age, breastfeeding is vital, along with fluid replacement (8). Many countries have introduced low-osmolarity ORS plus zinc for the treatment of cholera.

TABLE 2.

Compositions of different rehydration solutionsa

Solution Content (mmol/L)
Na+ K+ Cl HCO3 Carbohydrate
i.v. lactated Ringer’s solution 130 4 109 28
i.v. normal saline, 0.9% 154 0 154 0
i.v. cholera saline (Dhaka solution) 133 13 98 48 140
ORS (standard) 90 20 80 10 (citrate) 111
ORS (WHO 2002)a 75 20 65 10 (citrate) 75 (glucose)
Rice-based ORS (e.g., Cera ORS 75) 75 20 65 10 (citrate) 27 g rice syrup solids
a

The data on the compositions of rehydration solutions are adapted from standard guidelines (56, 58, 145).

FIG 3.

FIG 3

Management of cholera based on the severity of dehydration. These guidelines have been adapted from WHO guidelines for the treatment of diarrhea (58).

Treatment with i.v. fluids can be considered if vomiting continues more than three times in 1 h or if ORS is not improving the patient’s condition. Instant corrections of fluid and electrolyte deficits are the bases of rehydration therapy in order to counterbalance ongoing losses. Severe cholera cases require i.v. fluid (25). The WHO recommends Ringer’s lactate solution (Na+ at 130 mmol/L, K+ at 4 mmol/L, Cl at 109 mmol/L, HCO3 at 28 mmol/L, and Ca+ at 1.5 mmol/L [osmolarity of 273 mmol/L and pH of 6.5]) over normal saline (Na+ at 154 mmol/L and K+ at 154 mmol/L [osmolarity of 308 mmol/L and pH of 4.5]) as it contains more potassium and bicarbonate. The International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), and other governing bodies of countries where cholera is endemic prefer to use the “Dhaka solution” or “cholera saline” (Na+ at 133 mmol/L, K+ at 13 mmol/L, Cl at 154 mmol/L, HCO3 at 48 mmol/L, carbohydrate at 140 mmol/L) (Table 2), which contains glucose as well as more potassium and bicarbonate than Ringer’s lactate solution and can reduce adverse outcomes such as electrolyte imbalances (58, 59).

Severely dehydrated patients need an instant i.v. fluid infusion at a bolus dose of 100 mL/kg of body weight over 3 h and a one-third infusion in the first 30 min (59). Patients aged 1 year and above need 30 mL/kg in the first 30 min and 70 mL/kg in the next 2 1/2 h. The total duration of rehydration is 6 h for children less than 1 year of age (Fig. 3). Severe cholera patients should be kept on a cholera cot (e.g., a cot with a hole and an underlying bucket) to monitor the ongoing fluid loss so that the actual amount of fluid can be replaced. When a patient is able to drink, ORS is started again for hydration (5). For all patients, the fluid infusion should be given in repetitive measures if danger signs (hypovolemia, low radial pulse, and deep breathing, etc.) appear even after starting an i.v. infusion as a bolus therapy (Fig. 4). Malnourished children require a high-energy diet after correction of fluid deficiency to prevent hypoglycemia, hyponatremia, and hypokalemia (58).

FIG 4.

FIG 4

Flow diagram of the course of a cholera patient from admission to recovery.

Antibiotics and Antimicrobial Resistance

The WHO has recommended antibiotics for severe cholera patients irrespective of age and for patients who require hospitalization (5, 60). Several studies have shown that antibiotics shorten the length of infectious diarrhea (from 5 days to 1 to 2 days) as well as decrease the volume of stool output by up to 50% (1, 43, 61). Tetracycline, fluoroquinolones, co-trimoxazole, doxycycline, ciprofloxacin, trimethoprim-sulfamethoxazole, erythromycin, and azithromycin are the most commonly used antimicrobials for treating cholera patients, but resistance has become a global concern (43). The choice of antibiotics depends on local antibiotic susceptibility patterns. In most countries, doxycycline is recommended as a first-line treatment as a 300-mg single oral dose for adults (including pregnant women) and as a 2- to 4-mg/kg single oral dose for children. If resistance to doxycycline is documented, azithromycin and ciprofloxacin are alternative options. For children under 12 years of age, azithromycin is prescribed as a 20-mg/kg (maximum, 1-g) single oral dose, and ciprofloxacin is prescribed as a 20-mg/kg (maximum, 1-g) single oral dose. A single oral dose of azithromycin (1 g) or ciprofloxacin (1 g) is prescribed to adults suffering from severe cholera (https://www.cdc.gov/cholera/treatment/antibiotic-treatment.html).

Antimicrobial resistance (AMR) against trimethoprim-sulfamethoxazole, nalidixic acid, furazolidone, tetracycline, and ciprofloxacin developed between 1990 and 2010 (62). Hence, frequent performance of antimicrobial testing on V. cholerae clinical isolates is recommended for the treatment of cholera patients in settings where cholera is endemic as multidrug resistance (MDR) has developed due to frequent chromosomal mutation. Over the past decade, whole-genome sequencing of V. cholerae isolates has shown mobile genetic elements (MGEs) from other bacterial species, plasmids, conjugative elements, superintegrons, and supplemental sequences, all of which could lead to AMR (63). Studies from Africa showed increased resistance of V. cholerae O1 serotype strains to co-trimoxazole, chloramphenicol, ampicillin, and tetracycline (64, 65). Countries such as India and Nepal have also reported V. cholerae AMR to nalidixic acid, ciprofloxacin, tetracycline, and furazolidone, and there is an increasing number of MDR V. cholerae El Tor strains (66, 67).

Between 2009 and 2014, 17% of V. cholerae strains were resistant to third-generation cephalosporins, and 93% MDR strains were detected, exhibiting resistance to streptomycin, nalidixic acid, tetracycline, and trimethoprim-sulfamethoxazole (68, 69). An alarming fact is that the isolates were also becoming resistant to ciprofloxacin and azithromycin as these antibiotics were the drugs of choice for the management of cholera cases (70).

Self-medication with antibiotics has been reported in 83% (120/144) of infected and symptomatic household contacts in Bangladesh (71). Antibiotics are easily obtainable without a doctor’s prescription, and the habit of frequent and incomplete courses is quite common in developing countries (72, 73). Most recently, a randomized controlled trial of electronic decision-based diarrheal management through mobile phones using the mHealth Diarrhea Management (mHDM) platform in 10 district hospitals in Bangladesh showed a 10% reduction in the ordering of antibiotics in hospitalized patients compared to paper-based decisions in clinical facilities. Additionally, that study also found a remarkable decrease in prescribing of nonindicated antibiotics using mHDM among patients <5 years (28.5%) and >18 years (11.8%) of age (74). Such findings may help with antimicrobial stewardship in Bangladesh.

V. cholerae susceptibility to antibiotics is usually tested by two methods, disk diffusion and MIC methods for antibacterial properties (75, 76). To better understand the pattern of declining sensitivity of V. cholerae to antibiotics and its alarming resistance surge in the world, several molecular techniques are being conducted globally. National surveillance systems to identify changing sensitivity patterns are being used in order to identify the most appropriate drugs for cholera management (77). Moreover, the advantage of stool culture with sensitivity tests relative to PCR or RDTs is the added ability to perform antimicrobial susceptibility testing (AST) and monitor resistance patterns.

Antibiotic Prophylaxis

Findings from a systematic review have revealed that household contacts of cholera cases obtained protection against the disease when antibiotics were given to them as prophylaxis (78). In another study, one dose of doxycycline (300 mg) was administered orally to all prisoners and prison staff to prevent the spread of cholera in a prison in Cameroon in 2004, and no new cholera cases were reported over the next 4 months (79). However, mass chemoprophylaxis with an antibiotic is not recommended by the WHO for cholera control as it can lead to antibiotic resistance, but selective chemoprophylaxis may be provided for the prophylactic treatment of close contacts of a cholera patient (80). More studies are needed to inform the future use of targeted chemoprophylaxis in high-risk subpopulations.

Micronutrients

Zinc supplementation in children <5 years of age can also reduce the length and stool volume of diarrhea. Studies in various countries have shown that the addition of zinc to ORS reduces the severity of diarrhea and subsequently limits the use of antimicrobials (81). Zinc inhibits basolateral potassium channels by blocking cAMP-induced chloride-dependent fluid secretion. Zinc also regenerates the intestinal epithelium and increases the secretion of enzymes and the absorption of water and electrolytes, thus enhancing the immune response (82, 83). Vitamin A supplementation is suggested for children 6 months to 5 years of age to avert further occurrences of diarrhea (84). A high-calorie diet may reduce hypokalemia, hypoglycemia, and malnutrition, even when diarrhea is present (85).

FUTURE TREATMENTS

Supplementation with probiotics is relatively new for cholera management (86). Cholera toxin alters the gut microbiota (87). Probiotics can restore the gut microbiome and thus can potentially be used to clinically manage cholera. The use of probiotics in cholera management may limit AMR by reducing antibiotic use. Studies have shown the important contribution of the gut microbiome to fighting cholera and other diarrheal diseases (82, 83). There are several bacterial species in the gut that have been found to suppress cholera infection. Ruminococcus obeum has a positive correlation with cholera recovery (88). Another study showed that Lactobacillus rhamnosus strain GG (ATCC 53103) and Bifidobacterium longum 46 (DSM 14583) in coculture with V. cholerae were capable of removing CT from its environment (89). The therapeutic use of lytic bacteriophages, which is known as “phage therapy,” is another novel approach to the treatment of cholera (90). Phages are efficient at killing MDR bacteria. Studies conducted in infant mice and rabbits found that a combination of three isolated V. cholerae-specific virulent phages (ICP1, ICP2, and ICP3) was able to decrease the bacterial load of V. cholerae and prevent cholera-like diarrhea (91). Experimental studies targeting the inhibitor of cystic fibrosis channel transmembrane (CFTR), inhibitors of virulence factors, and a monosialoganglioside (GM1) antagonist found that they reduced intestinal secretion induced by CT of V. cholerae (92).

INTEGRATED CONTROL

Water, Sanitation, and Hygiene

Water, sanitation, and hygiene (WaSH) are of the utmost importance for the prevention and control of cholera and other enteric infections in the developing world (88, 89). Treatment of water at the source will not prevent the disease as contamination may occur at any time from collection to consumption. Water storage can also lead to contamination (93). When an outbreak is declared, interdisciplinary action is crucial for containing the pathogen, along with prioritizing which components of WaSH need to be implemented (93). Behavioral change practices (e.g., handwashing, use of soapy water, chlorination of household drinking water) have been shown to improve the protective efficacy of oral cholera vaccination when administered together (94). The Philippines, Thailand, and Vietnam have initiated cholera control strategies comprising surveillance, health promotion, and the supply of safe water. But in the cross-border areas of Malaysia, great challenges remain where the scarcity of sterile water, poor hygiene, and open defecation are noticeable concerns. Vietnam has reported zero cholera cases since 2012, and the Philippines has implemented a zero-open-defecation program along with other WaSH interventions to control the disease (95). Countries with high cholera disease burdens, such as Bangladesh, Pakistan, Nepal, and India, have minimal piped water in many localities, limited access to potable water in rural areas, shared latrines among the urban poor, open defecation in rural areas, and poor compliance with handwashing (96). Reports on evidence of household water treatment reducing cholera incidence have been published, but it is not effective in high-risk households due to financial constraints, poor education, and practices resulting in the low uptake of interventions (96, 97). Few high-risk households (12% in Nigeria, 19% in Nepal, and 24% in Haiti) have reported using water treatment (e.g., filtration, boiling, UV purification, or chlorine disinfectant use) (97). Without the support of national and local governments and nongovernment organizations, such implementations are impossible to sustain. Additionally, effective sewage systems with safe waste disposal and mechanisms to prevent untreated waste from reentering the environment are essential for controlling cholera (97). A strategy implemented in Mozambique, following the impact of Cyclone Idai in March 2019, was successful in controlling an outbreak of cholera. This included the establishment of a real-time surveillance system, WaSH, and vaccination (98). The WHO also recommends reinforcement and access to improved potable water, standard sanitation laws for food industries, execution of handwashing practices with soap, and safe handling of food as part of the cholera prevention strategy (5).

Inexpensive WaSH interventions such as solar power water purifiers, handwashing facilities, soapy water dispensers, Aquatabs (hypochlorous acid) for water purification, and safe storage containers (Fig. 5) have been modestly effective in lowering the numbers of cholera cases in community trials (99). During outbreaks, emergency WaSH interventions, including inexpensive WaSH strategies, health education, and media coverage, are crucial for reducing mortality as well as preventing further outbreaks (99). Community engagement plays an important role in long-term behavior changes and the prevention of cholera. Awareness campaigns using advertisements on the radio, television, billboards, and text messages can be used to educate people on when to seek medical attention and the use of ORS.

FIG 5.

FIG 5

Low-cost WaSH interventions for the control of cholera during an outbreak.

Vaccination

In areas where cholera is endemic, the WHO recommends cholera vaccination as part of the national cholera control program along with WaSH (10). Several studies have shown that the early introduction of vaccines during an outbreak offers 79% protection against cholera (100, 101), and even one dose of the cholera vaccine significantly reduces the risk (5, 102104).

Broadly speaking, there are four different types of cholera vaccines (Table 3): (i) killed whole-cell (WC) monovalent V. cholerae O1 (classical Inaba, and Ogawa, and El Tor Inaba) vaccines with a recombinant B subunit of cholera toxin, (ii) killed WC modified bivalent V. cholerae O1 and V. cholerae O139 vaccines without the B subunit, (iii) live attenuated oral cholera vaccines (OCVs), and (iv) parenteral cholera vaccines (5, 105, 106). Presently, only two OCVs are available worldwide: (i) the killed WC monovalent vaccine with a recombinant B subunit of cholera toxin (Dukoral) and (ii) killed and modified WC (O1 and O139) vaccines without the B subunit (Shanchol, Euvichol, Euvichol Plus, and mORC-Vax [106]).

TABLE 3.

Comparison of oral cholera vaccinesa

Vaccine Description for vaccine
Dukoral OraVacs Shanchol Euvichol/Euvichol Plus mORC-Vax Cholvax Vaxchora Hillchol
Generic name WC-rBSb WC-rBSb Modified bivalent WC Modified bivalent WC Modified bivalent WC Modified bivalent WC CVD 103-HgR
Type Oral killed; buffer is needed Oral killed; capsule Oral killed; no buffer Oral killed; no buffer Oral killed; no buffer Oral killed; no buffer Oral live attenuated; buffer is needed Oral killed; no buffer
Composition Monovalent; killed whole-cell vaccine O1 serogroup (classical Inaba, and Ogawa, and El Tor Inaba) and recombinant cholera toxin B subunit Monovalent; killed whole-cell vaccine O1 serogroup (classical Inaba and El Tor Inaba) and recombinant cholera toxin B subunit Bivalent; killed modified whole-cell bivalent (O1 and O139) vaccine without the B subunit Bivalent; killed modified whole-cell bivalent (O1 and O139) vaccine without the B subunit Bivalent; killed modified whole-cell bivalent (O1 and O139) vaccine without the B subunit Bivalent; killed modified whole-cell bivalent (O1 and O139) vaccines without the B subunit Monovalent; live attenuated serogroup O1 classical Inaba strain 569B Single strain; whole-cell recombinant Hikojima strain MS1568
Age range (yrs) ≥2 ≥2 ≥1 ≥1 ≥1 ≥1 18–64 1–45
Dose regimen(s) 2 doses given 1–6 wks apart, 3 doses for children aged 2–5 yrs 2 doses, 14 days apart 2 doses, 14 days apart 2 doses, 14 days apart 3 doses, days 0, 7, and 28 Single dose 2 doses, 14 days apart
Manufacturer Developed by SBL, Sweden; now Valneva, France Shanghai United Cell Biotechnology, China Shantha Biotechnics, India Eubiologics, South Korea Vabiotech, Hanoi, Vietnam Incepta Vaccine Ltd., Bangladesh PaxVax Inc., USA Incepta Vaccine Ltd., Bangladesh
First licensing country(ies), yr(s) Sweden, 1991 China and Philippines India, 2009 South Korea, 2004 Vietnam, 1997, 2009 Bangladesh, 2019 USA, 2016 Under development
WHO prequalification date (mo and yr) October 2001 No September 2011 December 2014/2017 No No No No
a

Information on the different oral cholera vaccines is adapted from references 5 and 106.

b

Whole-cell recombinant cholera B subunit.

Killed whole-cell cholera vaccines.

(i) Monovalent WC V. cholerae O1 oral cholera vaccine with a recombinant B subunit of cholera toxin.

The killed WC monovalent OCV was first manufactured in Sweden and gained licensure in 1991. Currently, it is available in more than 60 countries (5). It is found under the trade name Dukoral, manufactured by Valneva, France, and achieved WHO prequalification in October 2001 (106). The vaccine was formulated in combination with a recombinant B subunit of cholera toxin and formalin- or heat-killed WC V. cholerae O1 (classical Inaba and Ogawa and El Tor Inaba). The B unit of V. cholerae toxin is analogous to the heat-labile toxin (LT) of enterotoxigenic E. coli (ETEC) in its composition and functional capability and elicits cross-protection against ETEC (5). The vaccine is administered as a three-dose regimen 1 week apart for those aged 2 to 6 years and as a two-dose regimen at least 1 week apart for individuals >6 years of age. The added B subunit in the vaccine can be neutralized by gastric acids, and hence, to protect its functionality, the vaccine is required to be administered along with a buffer solution. The Dukoral vaccine does not defend against the V. cholerae O139 serogroup or other types of vibrios and has also been reported to elicit higher immune responses in cholera-naive populations than in populations where cholera is endemic (5, 105, 106).

OraVacs is a monovalent, killed WC vaccine containing the O1 serogroup (classical or El Tor biotype) and a recombinant cholera toxin B (rCTB) subunit and is indicated for traveler’s diarrhea. A clinical trial of this vaccine revealed that it is safe and immunogenic for children older than 2 years of age and adults. However, no efficacy trial data are available (106). This vaccine is manufactured by Shanghai United Cell Biotechnology, China, and is licensed in China and the Philippines (Table 3).

(ii) Bivalent modified WC V. cholerae O1 and V. cholerae O139 vaccines.

In the mid-1980s, scientists in Vietnam developed a modified killed WC vaccine comprising the V. cholerae O1 serogroup excluding CTB, ORC-Vax, with technology transfer from Sweden (105), and including both V. cholerae O1 and O139; hence, this vaccine was called the bivalent killed WC vaccine and was first licensed in Vietnam in 1997 (5, 105, 106). It was manufactured by Vabiotech (Hanoi, Vietnam), and it was included in the national Expanded Program for Immunization (EPI) and used during cholera outbreaks in that country. However, this vaccine was not WHO prequalified as Vietnam’s National Regulatory Agency (NRA) was not recognized by the WHO (105). Hence, to comply with international guidelines, the vaccine’s manufacturing technology was transferred to the International Vaccine Institute (IVI) (Seoul, South Korea) in 2004 from Vabiotech. This reformulated vaccine was named mORC-Vax and is now licensed in Vietnam. Several clinical trials have evaluated the two-dose schedule of this reformulated vaccine and found it to be safe and to produce antibody responses to O1, but immunogenicity was less pronounced against the O139 serotype (107, 108).

The IVI transferred the modified technology to Shantha Biotechnics (Hyderabad, India) to distribute mORC-Vax internationally. The vaccine was reformulated once again to WHO standards and met the requirements of safety and immunogenicity following several clinical trials. The modified version of the vaccine was called Shanchol (Shantha Biotechnics-Sanofi Pasteur), and it was licensed in 2009 and subsequently WHO prequalified in 2011 (105, 106). This final version of the vaccine was found to have fewer adverse reactions and higher levels of antibody responses in India, Bangladesh, Vietnam, and Ethiopia (107110). In Kolkata, India, and Bangladesh, large phase II and III trials were conducted, and it was proven to be safe and efficacious. During the 2-, 3-, and 5-year follow-up periods, the vaccine provided 67%, 66%, and 65% protection against V. cholerae O1, respectively (111113). A single dose of the Shanchol OCV is also effective in individuals above 5 years of age in settings where cholera is highly endemic (114, 115). The vaccine is safe and stable at elevated temperatures and is presently stockpiled (116). To meet the increasing demand for the OCV internationally, the IVI transferred the vaccine manufacturing technology to Eubiologics (Seoul, South Korea) to manufacture Euvichol, similar to Shanchol. The Euvichol (glass vial) vaccine was licensed in 2004 by the Government of South Korea, and the vaccine was prequalified by the WHO in 2015 (117). Clinical trials were conducted in South Korea and the Philippines, and the study outcomes revealed that robust antibody responses after two doses of Euvichol were comparable to those elicited by Shanchol in adults (82% versus 76%) and children (87% versus 89%) (117). Euvichol Plus (plastic vial) was developed at a lower cost, is easier to store, and received WHO prequalification in 2017 (105, 106). Euvichol is now stockpiled.

Live attenuated oral cholera vaccine.

In the 1970s, researchers from the Center for Vaccine Development (CVD) at the University of Maryland School of Medicine manufactured a live attenuated OCV with a V. cholerae O1 strain (106, 118). The live attenuated oral vaccine (like other live vaccines) provides greater efficacy, a more rapid immune response, and greater long-term protection among naive populations than among populations in areas where cholera is endemic (106, 119). To date, four live attenuated oral cholera vaccines have been produced, one of which is currently available, CVD 103-HgR (119).

CVD 103-HgR is the first live attenuated single-dose OCV (monovalent classical Inaba O1). Commercially, this vaccine was produced by the Swiss Serum and Vaccine Institute (SSVI), Berne, with the market names Orochol and Mutacol. Unfortunately, these vaccines were withdrawn from the market in 2003 for economic reasons (120, 121). In 2016, PaxVax Inc., a pharmaceutical company in the United States, acquired the licensure of CVD 103-HgR with the market name Vaxchora for U.S. adult travelers (122). Single-dose administration of Vaxchora showed a >90% vibriocidal seroconversion rate and gave protection among U.S. adult study participants following vaccination (119, 120, 122, 123). It had 79% protective efficacy when administered during a mass cholera vaccination campaign following an epidemic in Micronesia (119). Moreover, recently, satisfactory antibody responses and very few adverse events were seen in U.S. adults, children, and HIV patients (119, 122).

Licensures of other live attenuated OCVs are currently under development, such as Peru-15, composed of the O1 El Tor Inaba strain (Cholera Garde; Harvard Medical School, USA); OCV VA 1.4 (Government of India); a live attenuated El Tor Ogawa strain (638); IEM 108 (China); and HaitiV (United States) (106, 124, 125).

Parenteral vaccines.

Several parenteral cholera vaccines have been developed, among which are a killed WC vaccine, a purified lipopolysaccharide vaccine, a killed WC vaccine in combination with different adjuvants, and a polysaccharide-cholera toxin conjugate vaccine (126, 127). Only the killed WC vaccine was broadly available for several years but was not recommended by the WHO as this vaccine provided only a short duration of protective efficacy (43% protection for 3 months), induced a high level of adverse events (106, 126, 127), and was not recommended for pregnant women (126, 128). Most recently, a preclinical study with a newly developed parenteral cholera conjugate vaccine composed of Inaba O-specific polysaccharide (OSP) and the recombinant tetanus toxoid showed significant boosting of vibriocidal immune responses in mice following a single dose (CVD 103-HgR) (129). The parenteral cholera vaccine does not protect against cholera caused by V. cholerae O139 and did not show great effectiveness during cholera outbreaks. It also has a high-adverse-event profile, especially after intramuscular (i.m.) or subcutaneous (s.c.) administration, including local pain, erythema, induration, fever, malaise, and headache in most individuals, in comparison to oral cholera vaccines. OCVs are also more feasible to administer in an emergency setting than any parenteral vaccine and are also safe for pregnant women (126).

New vaccines under development.

Incepta Vaccine Ltd., a Bangladeshi company, has developed two OCVs, Cholvax and Hillchol. The technology for Cholvax vaccine production was transferred from the IVI (106). Cholvax has the same formulation as that of the Shanchol vaccine regarding strains and other formulations required for maintaining international good manufacturing practice (GMP) standards and WHO production guidelines (130). The manufacturing process for the Cholvax vaccine is also less expensive than those for the other available WHO-prequalified OCVs, and Cholvax has been found to be safe, immunogenic, and noninferior to the Shanchol vaccine (106, 131). Cholvax has been approved by the Directorate General of Drug Administration (DGDA) and was licensed in Bangladesh in 2019. The annual capacity for the production of Cholvax is 20 million to 40 million doses, which will be helpful to reduce the cholera burden in Bangladesh (106).

Hillchol is a formaldehyde-inactivated WC single-strain vaccine that originated from recombinant Hikojima V. cholerae strain MS1568 and was generated from an El Tor V. cholerae O1 parent/ancestor Inaba strain (Phil6973). This strain contains 50% LPS of each of the Ogawa and Inaba serotypes (132). The new monovalent Hillchol vaccine is manufactured with a solitary inactivation process and is anticipated to have a lower price than other current OCVs. The Hillchol vaccine was found to be safe, immunogenic, and noninferior to Shanchol among study participants (healthy adults and older and younger children) in Bangladesh (133).

The amended heat-stable Hillchol-B vaccine is composed of WC, formalin-inactivated Ogawa and Inaba strains in combination with rCTB in an enteric-coated capsule and can be easily administered during cholera outbreaks and among travelers to areas where cholera is endemic. Very recently, this vaccine was named the DuoChol capsule (124). Another OCV is in preclinical development in Sweden and is composed of formalin-killed cocultured isogenic El Tor Ogawa and Inaba serogroups (112). Eubiologics and the IVI are developing a formalin-killed classical Ogawa (Cairo50) and El Tor Inaba (Phil6973) vaccine, which has completed preclinical trials and is now set to move forward with a clinical trial in South Korea (124).

Vaccine enhancement in vulnerable populations.

Many low-income countries where cholera is endemic have reported a high cholera prevalence among young children. Current vaccines have shown a minimal level of protection and a short duration of protection in those aged 2 to 5 years (10, 126). Many approaches have been suggested to improve vaccine efficacy for this age cohort. Different regimes of vaccine administration, such as 3 doses at 4-week intervals or 2 doses at 8-week intervals, have been suggested to enhance immunogenicity in children. One study showed that withholding breastfeeding 3 h before vaccination increases vaccine efficacy along with supplementation with 20 mg of zinc per day for 42 days (134). Blood group, gut microbiota, malnutrition, environmental enteropathy, and the presence of multiple copathogens may also have a strong association with lower immunity in children (119).

The WHO recommends OCVs for pregnant and lactating women as these vaccines have potential benefits that outweigh the negligible risks (5). During pregnancy, severe dehydration can lead to premature delivery, miscarriage, and fetal death. OCVs were found to be safe, with no adverse fetal outcomes observed in several studies (135138). The WHO now recommends the use of OCVs for pregnant mothers in areas where cholera is endemic to prevent severe dehydrating cholera that may harm the fetus.

Herd immunity and vaccines.

It has been shown that inactivated OCVs can give significant herd protection in various study settings, which were analyzed using geographic information system (GIS) tools (138). The herd-protective effects of OCVs were measured using various study designs (individually or cluster randomized trials and cohort or case-control studies), and significant herd protection (both direct and indirect) against cholera was seen among unvaccinated persons and in the community (138). Mathematical modeling of cholera transmission (139) using Matlab showed that 93% of cholera infections in Bangladesh can be prevented with 50% OCV coverage (140). The model predicts an 89% drop in the incidence of cholera in the population that is not vaccinated. Study findings in Zanzibar showed that after mass cholera vaccination, OCVs also gave herd immunity (both direct and indirect) in this African setting (101). A large feasibility trial in Bangladesh revealed that children less than 3 years of age had a 47% reduction in the incidence of cholera if their mothers were given OCVs (139). Another analysis by Ali et al. showed that the chance of having cholera in the unvaccinated adult population was reduced by 14% with a 10% rise in OCV coverage in all age strata with a killed oral vaccine (138).

Challenges with oral cholera vaccines.

In 2013, after the formation of the global cholera vaccine stockpile, the logistic complications of vaccine delivery and the consistent scarcity of the vaccine supply hampered successful OCV implementation. The main challenge is to deliver two doses of the vaccine at a 14-day interval in rural or urban field settings (141). Recently, a heat stability study conducted on Shanchol showed that the vaccine is thermostable (116). Fortunately, Shanchol can be used at an ambient temperature (up to 42°C) for up to 1 month, but the storage temperature should be between 2°C and 8°C according to WHO guidelines (5).

Future vaccine strategies.

A single dose of an OCV induces a vibriocidal response among exposed populations, as observed in previous clinical trials (109, 111, 114, 142). A single dose of an OCV was shown to be efficacious (57%) among those above 5 years of age; however, no protective efficacy was observed for those below 5 years of age (114). A possible reason is a lack of acquired immunity. Thus, the memory B cell response against cholera-specific antigen develops over time due to recurrent natural exposure to V. cholerae (106, 142).

Studies on the use of booster doses of OCVs have been carried out in Bangladesh, which showed that children who received a single dose of an OCV 3 years earlier showed significantly increased vibriocidal antibody responses after receiving one booster dose of an OCV compared to those who did not receive an OCV earlier (142). These results suggest that boosting with one dose of an OCV augmented the immune responses in children, although more studies are required to adjust the primary booster doses of OCVs as well as to determine the duration between the prime and booster doses (142). Nevertheless, one dose of an OCV was also found to be protective during an outbreak among people (5 years of age and older) in Zambia who had less exposure to cholera (100).

Large campaigns of two doses of an OCV were carried out in 2017 among the Rohingya population (forcibly displaced Myanmar Nationals) in Cox’s Bazar in Bangladesh. A trial was conducted to assess the immunological parameters before and after vaccination. The study revealed a significant increase in vibriocidal antibody titers 14 days following the first dose of the OCV (143). Similarly, another study conducted during a humanitarian crisis in South Sudan showed that only one dose of an OCV was immunogenic and induced short-term antibodies (106, 115).

CHOLERA ELIMINATION

The GTFCC of the WHO has launched an initiative, entitled Ending Cholera: a Global Roadmap to 2030, aiming for at least a 90% mortality reduction in 47 countries of endemicity. The global roadmap aligns health and WaSH resources and targets areas most in need, saving lives, enhancing equity, and reducing the significant economic burden of cholera as well as other enteric diseases. The global roadmap focuses on three strategic priorities to control cholera. The first strategy includes the rapid detection of cholera cases and early responses to outbreaks through an early-warning surveillance system (EWARS) with an enhanced laboratory culture capacity along with dedicated health care facilities to treat cholera. The second strategy focuses on averting cholera recurrence in identified hot spots by improving WaSH and the delivery of OCVs. The third strategy is to develop a well-organized and efficient network to provide financial support to countries where cholera is endemic and bring national and international collaborators together to promote intersectorial coordination, supply mobilization, technical assistance, and strong cooperation to control cholera (https://www.gtfcc.org/wp-content/uploads/2019/10/gtfcc-ending-cholera-a-global-roadmap-to-2030.pdf).

Bangladesh is one of 20 countries of endemicity targeting cholera elimination. The incidence rate for cholera is 1.64 per 1,000 population annually. Bangladesh formulated the National Cholera Control Plan (NCCP) (https://www.gtfcc.org/wp-content/uploads/2020/08/6th-annual-meeting-gtfcc-bangladesh.pdf) in 2019, which is the guiding document to ensure OCV delivery to the target population according to a GTFCC strategic approach. A large OCV campaign was demonstrated among 1.2 million people as part of the NCCP in February 2020 in the capital city of Dhaka, comprising six high-risk cholera-prone areas (https://www.dtnext.in/world/2020/02/20/bangladesh-begins-1st-nationwide-anticholera-drive).

Along with Bangladesh, Uganda, Zambia, and Zanzibar are also in the process of trying to meet the Ending Cholera: a Global Roadmap to 2030 objectives. The Zambia government is working toward improving the WaSH sector in compliance with the roadmap supported by the GTFCC (https://www.gtfcc.org/wp-content/uploads/2020/08/5th-gtfcc-2018-zambia-francis-bwalya.pdf). Zanzibar is implementing the Zanzibar Comprehensive Cholera Elimination Plan (ZACCEP) (https://www.gtfcc.org/wp-content/uploads/2019/05/national-cholera-plan-zanzibar.pdf), which is a 10-year program to eliminate the indigenous transmission of cholera and promote a healthy and clean environment. Uganda has incorporated a 5-year plan to reduce cholera by 50% by 2022 through vaccination and the implementation of WaSH among 300,000 persons in the first 3 years, as reported at the 5th annual meeting of the GTFCC (https://www.gtfcc.org/wp-content/uploads/2020/08/5th-gtfcc-2018-uganda-immaculate-ampaire.pdf). Developed countries have eliminated cholera largely through improved sewage systems and clean water supplies. However, in the 21st century, high-risk populations and those in LMICs still do not have access to safe drinking water and formal sewage systems (97).

CONCLUSIONS

V. cholerae causes periodic cholera epidemics in several regions around the globe. The disease requires immediate treatment as it can cause death within hours in patients with moderate to severe cholera. With the development of i.v. fluids, ORS, and Zn therapy, progression to severe dehydration and mortality has been remarkably reduced. Antibiotics, micronutrients, and probiotics can further assist in recovery. In the developing world, treatment challenges are primarily due to delays in receiving prompt medical attention by health care professionals. The use of cholera RDTs as a point-of-care diagnostic during an outbreak along with PCR requires significant laboratory investment and skilled personnel. The ambitious global roadmap to end cholera by 2030 requires countries of endemicity to use evidence-based solutions to make this goal a reality (https://www.gtfcc.org/wp-content/uploads/2019/10/gtfcc-ending-cholera-a-global-roadmap-to-2030.pdf). To control cholera in endemic or outbreak situations at the domestic and communal levels, an OCV is considered an essential component of an integrated control package along with WaSH. Extensive and robust cholera surveillance, rapid diagnostics, treatment, and health education will be required for sustained control. Finally, to make all of this possible there must be political will at all levels of government. Without such support, the WHO 2030 targets will not be met.

ACKNOWLEDGMENTS

This article received no funding from any donor agency in the public, profitable, or nonprofitable sector.

The icddr,b is thankful to the Governments of Bangladesh, Canada, Sweden, and the United Kingdom for providing core/unrestricted support.

There is no conflict of interest.

Biographies

graphic file with name cmr.00211-21-f006.gif

Fahima Chowdhury, Associate Scientist and Project Coordinator, Mucosal Immunology and Vaccinology Unit, Infectious Diseases Division, icddr,b, is currently doing her Ph.D. under the Faculty of Medicine at Griffith University, Australia, under the supervision of Professor Nigel McMillan, Professor Allen Ross, and Dr. Firdausi Qadri. Dr. Fahima is working on different immune response studies in cholera patients and recently conducted an immunogenicity trial among forcefully displaced Myanmar Nationals (FDMNs). She was actively involved in large field trials of oral cholera vaccines (OCVs). Currently, as a PI, she completed clinical trials of locally produced vaccines such as cholera and hepatitis B vaccines. Very recently, she has been conducting a study among hospitalized COVID-19 patients to detect virus-specific antibodies and measure the immune responses in vitro and in vivo as well as antibody responses after different COVID-19 vaccines. She had published around 95 papers in peer-reviewed journals, including 16 papers as a first author.

graphic file with name cmr.00211-21-f007.gif

Allen G. Ross is Professor of Medicine and Executive Director of the Rural Health Research Institute with Charles Sturt University, Orange, NSW, Australia. Professor Ross’s expertise and research interests lie in the realm of graduate education, global health, medical microbiology, tropical infectious diseases, enteric diseases, disease control, pandemic planning, and vaccination. Professor Ross completed a bachelor of science in Biology (B.Sc., 1990) and a master of Science (M.Sc., 1994) in Human Biology in Canada before proceeding to Brisbane, Australia, where he completed a doctorate in Tropical Health with distinction (Ph.D., 1998); a bachelor of Medicine, bachelor of Surgery (M.B.B.S., 2005); a doctorate of Medicine (M.D., 2010); and a doctorate of Medical Science (D.Sc., 2017). He is a Fellow of the Royal College of Physicians of Edinburgh in the United Kingdom, Fellow of the Royal College of Pathology in the United Kingdom, Fellow of the Australasian College of Tropical Medicine, and Fellow of the Royal Society of Public Health in the United Kingdom.

graphic file with name cmr.00211-21-f008.gif

Md Taufiqul Islam, Deputy Project Coordinator, Mucosal Immunology and Vaccinology Unit, Infectious Diseases Division, icddr,b, completed his M.B.B.S. from Mymensingh Medical College Hospital, University of Dhaka, in 2008. Subsequently, he obtained his M.P.H. degree in 2016. He was enrolled in the Ph.D. program at Griffith University, Australia, in 2020. His area of work includes the introduction of an oral cholera vaccine in Bangladesh and a single-dose oral cholera vaccine study in urban Dhaka. Dr. Islam has contributed to preparing the Cholera Vaccine Investment Strategy, which is an essential instrument to enhance the cholera control program in Bangladesh. Evaluation of the safety of using whole-cell oral cholera vaccines in pregnant women is another important scientific involvement. Moreover, his area of work has been expanded by being involved in the vaccine delivery and surveillance network in the complex humanitarian crisis in Bangladesh. He is also actively working in the program for the control of endemic cholera in Bangladesh to achieve the target of End Cholera 2030, the global roadmap.

graphic file with name cmr.00211-21-f009.gif

Nigel A. J. McMillan, Griffith Centre for Cell and Gene Medicine, Menzies Health Institute, Queensland, Griffith University, obtained his Ph.D. at the University of Otago, New Zealand, in 1991. He is currently Professor of Infectious Diseases at Griffith University and explores the links between viruses and cancer. This includes developing novel nanoparticle delivery systems for RNAi and CRISPR. His laboratory was the first to report a cure for cancer using CRISPR-loaded nanoparticles in 2019, and in 2021, his laboratory published the first cure for SARS-CoV-2 infection in animal models using siRNA.

graphic file with name cmr.00211-21-f010.gif

Firdausi Qadri, Ph.D., is a Senior Scientist, Infectious Diseases Division, at icddr,b, Dhaka, Bangladesh. She also heads the Mucosal Immunology and Vaccinology Unit. Her work includes basic and applied immunology of infectious diseases but also clinical and large field-based studies on enteric vaccines and has coauthored 406 peer-reviewed publications, including reviews and book chapters. Special interests are infections caused by V. cholerae, Shigella, ETEC, Salmonella spp., and Helicobacter pylori under grants from Sida, the NIH, BMGF, USAID, the University of Oxford, and the European Union. The results have produced an impact on the field of enteric diseases, specifically in the areas of immunological, genetic, and genomic mechanisms, proteomics, metagenomics, diagnostics, and vaccine development. She has been elected fellow of many societies, including ASM, AAM, TWAS, IDSA, BAS, and INSA, and serves on advisory boards, including the ISDB science, biotechnology, and innovation board.

REFERENCES

  • 1.Sharifi-Mood B, Metanat M. 2014. Diagnosis, clinical management, prevention, and control of cholera; a review study. Int J Infect 1:e18303. 10.17795/iji-18303. [DOI] [Google Scholar]
  • 2.Clemens JD, Nair GB, Ahmed T, Qadri F, Holmgren J. 2017. Cholera. Lancet 390:1539–1549. 10.1016/S0140-6736(17)30559-7. [DOI] [PubMed] [Google Scholar]
  • 3.Deen J, Mengel MA, Clemens JD. 2020. Epidemiology of cholera. Vaccine 38:A31–A40. 10.1016/j.vaccine.2019.07.078. [DOI] [PubMed] [Google Scholar]
  • 4.Richterman A, Sainvilien DR, Eberly L, Ivers LC. 2018. Individual and household risk factors for symptomatic cholera infection: a systematic review and meta-analysis. J Infect Dis 218:S154–S164. 10.1093/infdis/jiy444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.World Health Organization. 2017. Cholera vaccines: WHO position paper—August 2017. Wkly Epidemiol Rec 92:477–500.28845659 [Google Scholar]
  • 6.Mandal S, Mandal MD, Pal NK. 2011. Cholera: a great global concern. Asian Pac J Trop Med 4:573–580. 10.1016/S1995-7645(11)60149-1. [DOI] [PubMed] [Google Scholar]
  • 7.Morris JG. 2011. Cholera—modern pandemic disease of ancient lineage. Emerg Infect Dis 17:2099–2104. 10.3201/eid1711.111109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Saha A, Rosewell A, Hayen A, MacIntyre CR, Qadri F. 2017. Improving immunization approaches to cholera. Expert Rev Vaccines 16:235–248. 10.1080/14760584.2017.1249470. [DOI] [PubMed] [Google Scholar]
  • 9.Goel AK, Jiang SC. 2010. Genetic determinants of virulence, antibiogram and altered biotype among the Vibrio cholerae O1 isolates from different cholera outbreaks in India. Infect Genet Evol 10:814–818. 10.1016/j.meegid.2009.06.022. [DOI] [PubMed] [Google Scholar]
  • 10.World Health Organization. 2010. Cholera vaccines: WHO position paper—March 2010. Wkly Epidemiol Rec 85:117–128. [PubMed] [Google Scholar]
  • 11.World Health Organization. 2017. Cholera, 2016. Wkly Epidemiol Rec 92:521–536.28884993 [Google Scholar]
  • 12.World Health Organization. 2018. Cholera, 2017. Wkly Epidemiol Rec 93:489–500. [Google Scholar]
  • 13.World Health Organization. 2019. Cholera, 2018. Wkly Epidemiol Rec 94:561–580. [Google Scholar]
  • 14.World Health Organization. 2020. Cholera, 2019. Wkly Epidemiol Rec 95:441–448. [Google Scholar]
  • 15.Harris JB, LaRocque RC, Qadri F, Ryan ET, Calderwood SB. 2012. Cholera. Lancet 379:2466–2476. 10.1016/S0140-6736(12)60436-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Qadri F, Svennerholm A-M, Faruque ASG, Sack RB. 2005. Enterotoxigenic Escherichia coli in developing countries: epidemiology, microbiology, clinical features, treatment, and prevention. Clin Microbiol Rev 18:465–483. 10.1128/CMR.18.3.465-483.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Alam M, Islam A, Bhuiyan NA, Rahim N, Hossain A, Khan GY, Ahmed D, Watanabe H, Izumiya H, Faruque ASG, Akanda AS, Islam S, Sack RB, Huq A, Colwell RR, Cravioto A. 2011. Clonal transmission, dual peak, and off-season cholera in Bangladesh. Infect Ecol Epidemiol 1:7273. 10.3402/iee.v1i0.7273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Akanda AS, Jutla AS, Islam S. 2009. Dual peak cholera transmission in Bengal Delta: a hydroclimatological explanation. Geophys Res Lett 36:L19401. 10.1029/2009GL039312. [DOI] [Google Scholar]
  • 19.Pascual M, Bouma MJ, Dobson AP. 2002. Cholera and climate: revisiting the quantitative evidence. Microbes Infect 4:237–245. 10.1016/s1286-4579(01)01533-7. [DOI] [PubMed] [Google Scholar]
  • 20.Lesmana M, Subekti D, Simanjuntak CH, Tjaniadi P, Campbell JR, Oyofo BA. 2001. Vibrio parahaemolyticus associated with cholera-like diarrhea among patients in North Jakarta, Indonesia. Diagn Microbiol Infect Dis 39:71–75. 10.1016/s0732-8893(00)00232-7. [DOI] [PubMed] [Google Scholar]
  • 21.Mukandavire Z, Morris JG. 2015. Modeling the epidemiology of cholera to prevent disease transmission in developing countries. Microbiol Spectr 3:VE-0011-2014. 10.1128/microbiolspec.VE-0011-2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.O’Connor KA, Cartwright E, Loharikar A, Routh J, Gaines J, Fouché M-DB, Jean-Louis R, Ayers T, Johnson D, Tappero JW, Roels TH, Archer WR, Dahourou GA, Mintz E, Quick R, Mahon BE. 2011. Risk factors early in the 2010 cholera epidemic, Haiti. Emerg Infect Dis 17:2136–2138. 10.3201/eid1711.110810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Glass RI, Holmgren J, Haley CE, Khan MR, Svennerholm AM, Stoll BJ, Belayet Hossain KM, Black RE, Yunus M, Barua D. 1985. Predisposition for cholera of individuals with O blood group. Possible evolutionary significance. Am J Epidemiol 121:791–796. 10.1093/oxfordjournals.aje.a114050. [DOI] [PubMed] [Google Scholar]
  • 24.Harris JB, Khan AI, LaRocque RC, Dorer DJ, Chowdhury F, Faruque ASG, Sack DA, Ryan ET, Qadri F, Calderwood SB. 2005. Blood group, immunity, and risk of infection with Vibrio cholerae in an area of endemicity. Infect Immun 73:7422–7427. 10.1128/IAI.73.11.7422-7427.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Larocque RC, Sabeti P, Duggal P, Chowdhury F, Khan AI, Lebrun LM, Harris JB, Ryan ET, Qadri F, Calderwood SB. 2009. A variant in long palate, lung and nasal epithelium clone 1 is associated with cholera in a Bangladeshi population. Genes Immun 10:267–272. 10.1038/gene.2009.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Karlsson EK, Harris JB, Tabrizi S, Rahman A, Shlyakhter I, Patterson N, O’Dushlaine C, Schaffner SF, Gupta S, Chowdhury F, Sheikh A, Shin OS, Ellis C, Becker CE, Stuart LM, Calderwood SB, Ryan ET, Qadri F, Sabeti PC, Larocque RC. 2013. Natural selection in a Bangladeshi population from the cholera-endemic Ganges River Delta. Sci Transl Med 5:192ra86. 10.1126/scitranslmed.3006338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Bavishi C, Dupont HL. 2011. Systematic review: the use of proton pump inhibitors and increased susceptibility to enteric infection. Aliment Pharmacol Ther 34:1269–1281. 10.1111/j.1365-2036.2011.04874.x. [DOI] [PubMed] [Google Scholar]
  • 28.Nsagha DS, Atashili J, Fon PN, Tanue EA, Ayima CW, Kibu OD. 2015. Assessing the risk factors of cholera epidemic in the Buea Health District of Cameroon. BMC Public Health 15:1128. 10.1186/s12889-015-2485-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Leung DT, Chowdhury F, Calderwood SB, Qadri F, Ryan ET. 2012. Immune responses to cholera in children. Expert Rev Anti Infect Ther 10:435–444. 10.1586/eri.12.23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Glass RI, Svennerholm AM, Stoll BJ, Khan MR, Hossain KM, Huq MI, Holmgren J. 1983. Protection against cholera in breast-fed children by antibodies in breast milk. N Engl J Med 308:1389–1392. 10.1056/NEJM198306093082304. [DOI] [PubMed] [Google Scholar]
  • 31.Chakraborty S, Deokule JS, Garg P, Bhattacharya SK, Nandy RK, Nair GB, Yamasaki S, Takeda Y, Ramamurthy T. 2001. Concomitant infection of enterotoxigenic Escherichia coli in an outbreak of cholera caused by Vibrio cholerae O1 and O139 in Ahmedabad, India. J Clin Microbiol 39:3241–3246. 10.1128/JCM.39.9.3241-3246.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Rinaldo A, Bertuzzo E, Blokesch M, Mari L, Gatto M. 2017. Modeling key drivers of cholera transmission dynamics provides new perspectives for parasitology. Trends Parasitol 33:587–599. 10.1016/j.pt.2017.04.002. [DOI] [PubMed] [Google Scholar]
  • 33.Waldor MK, Edward TR. 2014. Vibrio cholerae, p 2471–2479. In Bennett JE, Dolin R, Blaser MJ (ed), Mandell, Douglas, and Bennett’s principles and practice of infectious diseases, 8th ed. Elsevier, Philadelphia, PA. [Google Scholar]
  • 34.Lebens M, Karlsson SL, Källgård S, Blomquist M, Ekman A, Nygren E, Holmgren J. 2011. Construction of novel vaccine strains of Vibrio cholerae co-expressing the Inaba and Ogawa serotype antigens. Vaccine 29:7505–7513. 10.1016/j.vaccine.2011.06.121. [DOI] [PubMed] [Google Scholar]
  • 35.Nandi B, Nandy RK, Vicente ACP, Ghose AC. 2000. Molecular characterization of a new variant of toxin-coregulated pilus protein (TcpA) in a toxigenic non-O1/non-O139 strain of Vibrio cholerae. Infect Immun 68:948–952. 10.1128/IAI.68.2.948-952.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Merrell DS, Butler SM, Qadri F, Dolganov NA, Alam A, Cohen MB, Calderwood SB, Schoolnik GK, Camilli A. 2002. Host-induced epidemic spread of the cholera bacterium. Nature 417:642–645. 10.1038/nature00778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Christaki E, Dimitriou P, Pantavou K, Nikolopoulos GK. 2020. The impact of climate change on cholera: a review on the global status and future challenges. Atmosphere 11:449. 10.3390/atmos11050449. [DOI] [Google Scholar]
  • 38.Sack DA, Sack RB, Nair GB, Siddique AK. 2004. Cholera. Lancet 363:223–233. 10.1016/S0140-6736(03)15328-7. [DOI] [PubMed] [Google Scholar]
  • 39.Azman AS, Rudolph KE, Cummings DAT, Lessler J. 2013. The incubation period of cholera: a systematic review. J Infect 66:432–438. 10.1016/j.jinf.2012.11.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ramamurthy T, Das B, Chakraborty S, Mukhopadhyay AK, Sack DA. 2020. Diagnostic techniques for rapid detection of Vibrio cholerae O1/O139. Vaccine 38:A73–A82. 10.1016/j.vaccine.2019.07.099. [DOI] [PubMed] [Google Scholar]
  • 41.Bhandari M, Jennison AV, Rathnayake IU, Huygens F. 2021. Evolution, distribution and genetics of atypical Vibrio cholerae—a review. Infect Genet Evol 89:104726. 10.1016/j.meegid.2021.104726. [DOI] [PubMed] [Google Scholar]
  • 42.Weil AA, Ivers LC, Harris JB. 2012. Cholera: lessons from Haiti and beyond. Curr Infect Dis Rep 14:1–8. 10.1007/s11908-011-0221-9. [DOI] [PubMed] [Google Scholar]
  • 43.Davies HG, Bowman C, Luby SP. 2017. Cholera—management and prevention. J Infect 74:S66–S73. 10.1016/S0163-4453(17)30194-9. [DOI] [PubMed] [Google Scholar]
  • 44.Harris JB, Ivers LC, Ferraro MJ. 2011. Case records of the Massachusetts General Hospital. Case 19-2011. A 4-year-old Haitian boy with vomiting and diarrhea. N Engl J Med 364:2452–2461. 10.1056/NEJMcpc1100927. [DOI] [PubMed] [Google Scholar]
  • 45.D’Mello-Guyett L, Gallandat K, Van den Bergh R, Taylor D, Bulit G, Legros D, Maes P, Checchi F, Cumming O. 2020. Prevention and control of cholera with household and community water, sanitation and hygiene (WASH) interventions: a scoping review of current international guidelines. PLoS One 15:e0226549. 10.1371/journal.pone.0226549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ryan ET, Dhar U, Khan WA, Salam MA, Faruque AS, Fuchs GJ, Calderwood SB, Bennish ML. 2000. Mortality, morbidity, and microbiology of endemic cholera among hospitalized patients in Dhaka, Bangladesh. Am J Trop Med Hyg 63:12–20. 10.4269/ajtmh.2000.63.12. [DOI] [PubMed] [Google Scholar]
  • 47.Ganesan D, Gupta SS, Legros D. 2020. Cholera surveillance and estimation of burden of cholera. Vaccine 38:A13–A17. 10.1016/j.vaccine.2019.07.036. [DOI] [PubMed] [Google Scholar]
  • 48.Huq A, Grim C, Colwell RR, Nair GB. 2006. Detection, isolation, and identification of Vibrio cholerae from the environment. Curr Protoc Microbiol Chapter 6:Unit 6A.5. 10.1002/9780471729259.mc06a05s02. [DOI] [PubMed] [Google Scholar]
  • 49.LaRocque RC, Harris JB. 2018. Cholera: clinical features, diagnosis, treatment, and prevention. In Calderwood SB (ed), UpToDate. Wolters Kluwer, Alphen aan den Rijn, The Netherlands. [Google Scholar]
  • 50.Benenson AS, Islam MR, Greenough WB, III.. 1964. Rapid identification of Vibrio cholerae by darkfield microscopy. Bull World Health Organ 30:827–831. [PMC free article] [PubMed] [Google Scholar]
  • 51.Nelson EJ, Chowdhury A, Harris JB, Begum YA, Chowdhury F, Khan AI, LaRocque RC, Bishop AL, Ryan ET, Camilli A, Qadri F, Calderwood SB. 2007. Complexity of rice-water stool from patients with Vibrio cholerae plays a role in the transmission of infectious diarrhea. Proc Natl Acad Sci USA 104:19091–19096. 10.1073/pnas.0706352104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Mehrabadi JF, Morsali P, Nejad HR, Imani Fooladi AA. 2012. Detection of toxigenic Vibrio cholerae with new multiplex PCR. J Infect Public Health 5:263–267. 10.1016/j.jiph.2012.02.004. [DOI] [PubMed] [Google Scholar]
  • 53.Harris JR, Cavallaro EC, de Nóbrega AA, Barrado JCBDS, Bopp C, Parsons MB, Djalo D, Fonseca FGDS, Ba U, Semedo A, Sobel J, Mintz ED. 2009. Field evaluation of Crystal VC rapid dipstick test for cholera during a cholera outbreak in Guinea-Bissau. Trop Med Int Health 14:1117–1121. 10.1111/j.1365-3156.2009.02335.x. [DOI] [PubMed] [Google Scholar]
  • 54.Islam MT, Khan AI, Sayeed MA, Amin J, Islam K, Alam N, Sultana N, Jahan N, Rashid MM, Khan ZH, Zion MI, Afrad MH, Siddique SA, Khanam F, Begum YA, Islam MS, Qadri F. 2019. Field evaluation of a locally produced rapid diagnostic test for early detection of cholera in Bangladesh. PLoS Negl Trop Dis 13:e0007124. 10.1371/journal.pntd.0007124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Parvin I, Shahid ASMSB, Das S, Shahrin L, Ackhter MM, Alam T, Khan SH, Chisti MJ, Clemens JD, Ahmed T, Sack DA, Faruque ASG. 2021. Vibrio cholerae O139 persists in Dhaka, Bangladesh since 1993. PLoS Negl Trop Dis 15:e0009721. 10.1371/journal.pntd.0009721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Pulungsih SP, Punjabi NH, Rafli K, Rifajati A, Kumala S, Simanjuntak CH, Yuwono, Lesmana M, Subekti D, Sutoto, Fontaine O. 2006. Standard WHO-ORS versus reduced-osmolarity ORS in the management of cholera patients. J Health Popul Nutr 24:107–112. [PubMed] [Google Scholar]
  • 57.Ramakrishna BS, Venkataraman S, Srinivasan P, Dash P, Young GP, Binder HJ. 2000. Amylase-resistant starch plus oral rehydration solution for cholera. N Engl J Med 342:308–313. 10.1056/NEJM200002033420502. [DOI] [PubMed] [Google Scholar]
  • 58.Department of Child and Adolescent Health and Development, World Health Organization. 2005. The treatment of diarrhoea: a manual for physicians and other senior health workers. Department of Child and Adolescent Health and Development, World Health Organization, Geneva, Switzerland. [Google Scholar]
  • 59.Pietroni MAC. 2020. Case management of cholera. Vaccine 38:A105–A109. 10.1016/j.vaccine.2019.09.098. [DOI] [PubMed] [Google Scholar]
  • 60.Nelson EJ, Nelson DS, Salam MA, Sack DA. 2011. Antibiotics for both moderate and severe cholera. N Engl J Med 364:5–7. 10.1056/NEJMp1013771. [DOI] [PubMed] [Google Scholar]
  • 61.Leibovici-Weissman Y, Neuberger A, Bitterman R, Sinclair D, Salam MA, Paul M. 2014. Antimicrobial drugs for treating cholera. Cochrane Database Syst Rev 2014:CD008625. 10.1002/14651858.CD008625.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Ghosh A, Ramamurthy T. 2011. Antimicrobials & cholera: are we stranded? Indian J Med Res 153:225–231. [PMC free article] [PubMed] [Google Scholar]
  • 63.Das B, Verma J, Kumar P, Ghosh A, Ramamurthy T. 2020. Antibiotic resistance in Vibrio cholerae: understanding the ecology of resistance genes and mechanisms. Vaccine 38:A83–A92. 10.1016/j.vaccine.2019.06.031. [DOI] [PubMed] [Google Scholar]
  • 64.Abera B, Bezabih B, Dessie A. 2010. Antimicrobial suceptibility [sic] of V. cholerae in north west, Ethiopia. Ethiop Med J 48:23–28. [PubMed] [Google Scholar]
  • 65.Mandomando I, Espasa M, Vallès X, Sacarlal J, Sigaúque B, Ruiz J, Alonso P. 2007. Antimicrobial resistance of Vibrio cholerae O1 serotype Ogawa isolated in Manhiça District Hospital, southern Mozambique. J Antimicrob Chemother 60:662–664. 10.1093/jac/dkm257. [DOI] [PubMed] [Google Scholar]
  • 66.Mandal J, Dinoop KP, Parija SC. 2012. Increasing antimicrobial resistance of Vibrio cholerae O1 biotype El Tor strains isolated in a tertiary-care centre in India. J Health Popul Nutr 30:12–16. 10.3329/jhpn.v30i1.11270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Rijal N, Acharya J, Adhikari S, Upadhaya BP, Shakya G, Kansakar P, Rajbhandari P. 2019. Changing epidemiology and antimicrobial resistance in Vibrio cholerae: AMR surveillance findings (2006-2016) from Nepal. BMC Infect Dis 19:801. 10.1186/s12879-019-4432-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Faruque ASG, Alam K, Malek MA, Khan MGY, Ahmed S, Saha D, Khan WA, Nair GB, Salam MA, Luby SP, Sack DA. 2007. Emergence of multidrug-resistant strain of Vibrio cholerae O1 in Bangladesh and reversal of their susceptibility to tetracycline after two years. J Health Popul Nutr 25:241–243. [PMC free article] [PubMed] [Google Scholar]
  • 69.Ceccarelli D, Alam M, Huq A, Colwell RR. 2016. Reduced susceptibility to extended-spectrum β-lactams in Vibrio cholerae isolated in Bangladesh. Front Public Health 4:231. 10.3389/fpubh.2016.00231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Rashed SM, Hasan NA, Alam M, Sadique A, Sultana M, Hoq MM, Sack RB, Colwell RR, Huq A. 2017. Vibrio cholerae O1 with reduced susceptibility to ciprofloxacin and azithromycin isolated from a rural coastal area of Bangladesh. Front Microbiol 8:252. 10.3389/fmicb.2017.00252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Weil AA, Khan AI, Chowdhury F, LaRocque RC, Faruque ASG, Ryan ET, Calderwood SB, Qadri F, Harris JB. 2009. Clinical outcomes in household contacts of patients with cholera in Bangladesh. Clin Infect Dis 49:1473–1479. 10.1086/644779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Biswas M, Roy MN, Manik MIN, Hossain MS, Tapu STA, Moniruzzaman M, Sultana S. 2014. Self medicated antibiotics in Bangladesh: a cross-sectional health survey conducted in the Rajshahi City. BMC Public Health 14:847. 10.1186/1471-2458-14-847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Larson CP, Saha UR, Islam R, Roy N. 2006. Childhood diarrhoea management practices in Bangladesh: private sector dominance and continued inequities in care. Int J Epidemiol 35:1430–1439. 10.1093/ije/dyl167. [DOI] [PubMed] [Google Scholar]
  • 74.Khan AI, Mack JA, Salimuzzaman M, Zion MI, Sujon H, Ball RL, Maples S, Rashid MM, Chisti MJ, Sarker SA, Biswas D, Hossin R, Bardosh KL, Begum YA, Ahmed A, Pieri D, Haque F, Rahman M, Levine AC, Qadri F, Flora MS, Gurka MJ, Nelson EJ. 2020. Electronic decision support and diarrhoeal disease guideline adherence (mHDM): a cluster randomised controlled trial. Lancet Digit Health 2:e250–e258. 10.1016/S2589-7500(20)30062-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Tan CW, Rukayadi Y, Hasan H, Thung TY, Lee E, Rollon WD, Hara H, Kayali AY, Nishibuchi M, Radu S. 2020. Prevalence and antibiotic resistance patterns of Vibrio parahaemolyticus isolated from different types of seafood in Selangor, Malaysia. Saudi J Biol Sci 27:1602–1608. 10.1016/j.sjbs.2020.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Gupta P, Modgil V, Kant V, Kaur H, Narayan C, Mahindroo J, Verma R, Mohan B, Taneja N. 2022. Phenotypic and genotypic characterization of antimicrobial resistance in clinical isolates of Vibrio cholerae over a decade (2002–2016). Indian J Med Microbiol 40:24–29. 10.1016/j.ijmmb.2021.11.008. [DOI] [PubMed] [Google Scholar]
  • 77.Malla S, Dumre SP, Shakya G, Kansakar P, Rai B, Hossain A, Nair GB, Albert MJ, Sack D, Baker S, Rahman M, Antimicrobial Resistance Surveillance Programme Team, Nepal . 2014. The challenges and successes of implementing a sustainable antimicrobial resistance surveillance programme in Nepal. BMC Public Health 14:269. 10.1186/1471-2458-14-269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Reveiz L, Chapman E, Ramon-Pardo P, Koehlmoos TP, Cuervo LG, Aldighieri S, Chambliss A. 2011. Chemoprophylaxis in contacts of patients with cholera: systematic review and meta-analysis. PLoS One 6:e27060. 10.1371/journal.pone.0027060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Guévart É, Solle J, Noeske J, Amougou G, Mouangue A, Fouda AB. 2005. Mass antibiotic prophylaxis against cholera in the New Bell central prison in Douala during the 2004 epidemic. Sante 15:225–227. [PubMed] [Google Scholar]
  • 80.Global Task Force on Cholera Control. 2020. Technical note use of antibiotics for the treatment and control of cholera 2018. World Health Organization, Geneva, Switzerland. https://www.gtfcc.org/wp-content/uploads/2019/10/gtfcc-technical-note-on-use-of-antibiotics-for-the-treatment-of-cholera.pdf. Accessed 29 July 2020. [Google Scholar]
  • 81.Qadri F, Bhuiyan TR, Sack DA, Svennerholm A-M. 2013. Immune responses and protection in children in developing countries induced by oral vaccines. Vaccine 31:452–460. 10.1016/j.vaccine.2012.11.012. [DOI] [PubMed] [Google Scholar]
  • 82.Roy SK, Hossain MJ, Khatun W, Chakraborty B, Chowdhury S, Begum A, Mah-e-Muneer S, Shafique S, Khanam M, Chowdhury R. 2008. Zinc supplementation in children with cholera in Bangladesh: randomised controlled trial. BMJ 336:266–268. 10.1136/bmj.39416.646250.AE. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Bajait C, Thawani V. 2011. Role of zinc in pediatric diarrhea. Indian J Pharmacol 43:232–235. 10.4103/0253-7613.81495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Francis DK. 2011. Vitamin A supplementation for preventing death and illness in children 6 months to 5 years of age. Cochrane Database Syst Rev 2011:ED000016. 10.1002/14651858.ED000016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Chowdhury F, Khan AI, Faruque ASG, Ryan ET. 2010. Severe, acute watery diarrhea in an adult. PLoS Negl Trop Dis 4:e898. 10.1371/journal.pntd.0000898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Focareta A, Paton JC, Morona R, Cook J, Paton AW. 2006. A recombinant probiotic for treatment and prevention of cholera. Gastroenterology 130:1688–1695. 10.1053/j.gastro.2006.02.005. [DOI] [PubMed] [Google Scholar]
  • 87.Cho JY, Liu R, Macbeth JC, Hsiao A. 2021. The interface of Vibrio cholerae and the gut microbiome. Gut Microbes 13:1937015. 10.1080/19490976.2021.1937015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Hsiao A, Ahmed AMS, Subramanian S, Griffin NW, Drewry LL, Petri WA, Jr, Haque R, Ahmed T, Gordon JI. 2014. Members of the human gut microbiota involved in recovery from Vibrio cholerae infection. Nature 515:423–426. 10.1038/nature13738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Heikkilä JE, Nybom SMK, Salminen SJ, Meriluoto JAO. 2012. Removal of cholera toxin from aqueous solution by probiotic bacteria. Pharmaceuticals (Basel) 5:665–673. 10.3390/ph5060665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Hsueh BY, Waters CM. 2019. Combating cholera. F1000Res 8:F1000 Faculty Rev-589. 10.12688/f1000research.18093.1. [DOI] [Google Scholar]
  • 91.Reyes-Robles T, Dillard RS, Cairns LS, Silva-Valenzuela CA, Housman M, Ali A, Wright ER, Camilli A. 2018. Vibrio cholerae outer membrane vesicles inhibit bacteriophage infection. J Bacteriol 200:e00792-17. 10.1128/JB.00792-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Sousa FBM, Nolêto IRSG, Chaves LS, Pacheco G, Oliveira AP, Fonseca MMV, Medeiros JVR. 2020. A comprehensive review of therapeutic approaches available for the treatment of cholera. J Pharm Pharmacol 72:1715–1731. 10.1111/jphp.13344. [DOI] [PubMed] [Google Scholar]
  • 93.Farmer P, Almazor CP, Bahnsen ET, Barry D, Bazile J, Bloom BR, Bose N, Brewer T, Calderwood SB, Clemens JD, Cravioto A, Eustache E, Jérôme G, Gupta N, Harris JB, Hiatt HH, Holstein C, Hotez PJ, Ivers LC, Kerry VB, Koenig SP, LaRocque RC, Léandre F, Lambert W, Lyon E, Mekalanos JJ, Mukherjee JS, Oswald C, Pape J-W, Prosper AG, Rabinovich R, Raymonville M, Réjouit J-R, Ronan LJ, Rosenberg ML, Ryan ET, Sachs JD, Sack DA, Surena C, Suri AA, Ternier R, Waldor MK, Walton D, Weigel JL. 2011. Meeting cholera’s challenge to Haiti and the world: a joint statement on cholera prevention and care. PLoS Negl Trop Dis 5:e1145. 10.1371/journal.pntd.0001145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Qadri F, Ali M, Chowdhury F, Khan AI, Saha A, Khan IA, Begum YA, Bhuiyan TR, Chowdhury MI, Uddin MJ, Khan JAM, Chowdhury AI, Rahman A, Siddique SA, Asaduzzaman M, Akter A, Khan A, You YA, Siddik AU, Saha NC, Kabir A, Riaz BK, Biswas SK, Begum F, Unicomb L, Luby SP, Cravioto A, Clemens JD. 2015. Feasibility and effectiveness of oral cholera vaccine in an urban endemic setting in Bangladesh: a cluster randomised open-label trial. Lancet 386:1362–1371. 10.1016/S0140-6736(15)61140-0. [DOI] [PubMed] [Google Scholar]
  • 95.Ahmed MU, Baquilod M, Deola C, Tu ND, Anh DD, Grasso C, Gautam A, Hamzah WM, Heng S, Iamsirithaworn S, Kadim M, Kar SK, Le Thi Quynh M, Lopez AL, Lynch J, Memon I, Mengel M, Long VN, Pandey BD, Quadri F, Saadatian-Elahi M, Gupta SS, Sultan A, Sur D, Tan DQ, Ha HTT, Hein NT, Lan PT, Upreti SR, Endtz H, Ganguly NK, Legros D, Picot V, Nair GB. 2018. Cholera prevention and control in Asian countries. BMC Proc 12:62. 10.1186/s12919-018-0158-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Taylor DL, Kahawita TM, Cairncross S, Ensink JHJ. 2015. The impact of water, sanitation and hygiene interventions to control cholera: a systematic review. PLoS One 10:e0135676. 10.1371/journal.pone.0135676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Luby SP, Davis J, Brown RR, Gorelick SM, Wong THF. 2020. Broad approaches to cholera control in Asia: water, sanitation and handwashing. Vaccine 38:A110–A117. 10.1016/j.vaccine.2019.07.084. [DOI] [PubMed] [Google Scholar]
  • 98.Lequechane JD, Mahumane A, Chale F, Nhabomba C, Salomão C, Lameira C, Chicumbe S, Semá Baltazar C. 2020. Mozambique’s response to cyclone Idai: how collaboration and surveillance with water, sanitation and hygiene (WASH) interventions were used to control a cholera epidemic. Infect Dis Poverty 9:68. 10.1186/s40249-020-00692-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Ross AG, Rahman M, Alam M, Zaman K, Qadri F. 2020. Can we ‘WaSH’ infectious diseases out of slums? Int J Infect Dis 92:130–132. 10.1016/j.ijid.2020.01.014. [DOI] [PubMed] [Google Scholar]
  • 100.Ferreras E, Chizema-Kawesha E, Blake A, Chewe O, Mwaba J, Zulu G, Poncin M, Rakesh A, Page A-L, Stoitsova S, Voute C, Uzzeni F, Robert H, Serafini M, Matapo B, Eiros J-M, Quilici M-L, Pezzoli L, Azman AS, Cohuet S, Ciglenecki I, Malama K, Luquero FJ. 2018. Single-dose cholera vaccine in response to an outbreak in Zambia. N Engl J Med 378:577–579. 10.1056/NEJMc1711583. [DOI] [PubMed] [Google Scholar]
  • 101.Khatib AM, Ali M, von Seidlein L, Kim DR, Hashim R, Reyburn R, Ley B, Thriemer K, Enwere G, Hutubessy R, Aguado MT, Kieny M-P, Lopez AL, Wierzba TF, Ali SM, Saleh AA, Mukhopadhyay AK, Clemens J, Jiddawi MS, Deen J. 2012. Effectiveness of an oral cholera vaccine in Zanzibar: findings from a mass vaccination campaign and observational cohort study. Lancet Infect Dis 12:837–844. 10.1016/S1473-3099(12)70196-2. [DOI] [PubMed] [Google Scholar]
  • 102.Amani A, Tatang CA, Bayiha CN, Woung M, Ngo Bama S, Nangmo A, Mbang MA, Epee Douba E. 2021. A reactive vaccination campaign with single dose oral cholera vaccine (OCV) during a cholera outbreak in Cameroon. Vaccine 39:1290–1296. 10.1016/j.vaccine.2021.01.017. [DOI] [PubMed] [Google Scholar]
  • 103.Luquero FJ, Grout L, Ciglenecki I, Sakoba K, Traore B, Heile M, Diallo AA, Itama C, Page A-L, Quilici M-L, Mengel MA, Eiros JM, Serafini M, Legros D, Grais RF. 2014. Use of Vibrio cholerae vaccine in an outbreak in Guinea. N Engl J Med 370:2111–2120. 10.1056/NEJMoa1312680. [DOI] [PubMed] [Google Scholar]
  • 104.Ivers LC, Hilaire IJ, Teng JE, Almazor CP, Jerome JG, Ternier R, Boncy J, Buteau J, Murray MB, Harris JB, Franke MF. 2015. Effectiveness of reactive oral cholera vaccination in rural Haiti: a case-control study and bias-indicator analysis. Lancet Glob Health 3:e162–e168. 10.1016/S2214-109X(14)70368-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Lopez AL, Gonzales MLA, Aldaba JG, Nair GB. 2014. Killed oral cholera vaccines: history, development and implementation challenges. Ther Adv Vaccines 2:123–136. 10.1177/2051013614537819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Shaikh H, Lynch J, Kim J, Excler J-L. 2020. Current and future cholera vaccines. Vaccine 38:A118–A126. 10.1016/j.vaccine.2019.12.011. [DOI] [PubMed] [Google Scholar]
  • 107.Anh DD, Canh DG, Lopez AL, Thiem VD, Long PT, Son NH, Deen J, von Seidlein L, Carbis R, Han SH, Shin SH, Attridge S, Holmgren J, Clemens J. 2007. Safety and immunogenicity of a reformulated Vietnamese bivalent killed, whole-cell, oral cholera vaccine in adults. Vaccine 25:1149–1155. 10.1016/j.vaccine.2006.09.049. [DOI] [PubMed] [Google Scholar]
  • 108.Mahalanabis D, Lopez AL, Sur D, Deen J, Manna B, Kanungo S, von Seidlein L, Carbis R, Han SH, Shin SH, Attridge S, Rao R, Holmgren J, Clemens J, Bhattacharya SK. 2008. A randomized, placebo-controlled trial of the bivalent killed, whole-cell, oral cholera vaccine in adults and children in a cholera endemic area in Kolkata, India. PLoS One 3:e2323. 10.1371/journal.pone.0002323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Saha A, Chowdhury MI, Khanam F, Bhuiyan MS, Chowdhury F, Khan AI, Khan IA, Clemens J, Ali M, Cravioto A, Qadri F. 2011. Safety and immunogenicity study of a killed bivalent (O1 and O139) whole-cell oral cholera vaccine Shanchol, in Bangladeshi adults and children as young as 1 year of age. Vaccine 29:8285–8292. 10.1016/j.vaccine.2011.08.108. [DOI] [PubMed] [Google Scholar]
  • 110.Desai SN, Akalu Z, Teshome S, Teferi M, Yamuah L, Kim DR, Yang JS, Hussein J, Park JY, Jang MS, Mesganaw C, Taye H, Beyene D, Bedru A, Singh AP, Wierzba TF, Aseffa A. 2015. A randomized, placebo-controlled trial evaluating safety and immunogenicity of the killed, bivalent, whole-cell oral cholera vaccine in Ethiopia. Am J Trop Med Hyg 93:527–533. 10.4269/ajtmh.14-0683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Sur D, Lopez AL, Kanungo S, Paisley A, Manna B, Ali M, Niyogi SK, Park JK, Sarkar B, Puri MK, Kim DR, Deen JL, Holmgren J, Carbis R, Rao R, Nguyen TV, Donner A, Ganguly NK, Nair GB, Bhattacharya SK, Clemens JD. 2009. Efficacy and safety of a modified killed-whole-cell oral cholera vaccine in India: an interim analysis of a cluster-randomised, double-blind, placebo-controlled trial. Lancet 374:1694–1702. 10.1016/S0140-6736(09)61297-6. [DOI] [PubMed] [Google Scholar]
  • 112.Sur D, Kanungo S, Sah B, Manna B, Ali M, Paisley AM, Niyogi SK, Park JK, Sarkar B, Puri MK, Kim DR, Deen JL, Holmgren J, Carbis R, Rao R, Nguyen TV, Han SH, Attridge S, Donner A, Ganguly NK, Bhattacharya SK, Nair GB, Clemens JD, Lopez AL. 2011. Efficacy of a low-cost, inactivated whole-cell oral cholera vaccine: results from 3 years of follow-up of a randomized, controlled trial. PLoS Negl Trop Dis 5:e1289. 10.1371/journal.pntd.0001289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Bhattacharya SK, Sur D, Ali M, Kanungo S, You YA, Manna B, Sah B, Niyogi SK, Park JK, Sarkar B, Puri MK, Kim DR, Deen JL, Holmgren J, Carbis R, Dhingra MS, Donner A, Nair GB, Lopez AL, Wierzba TF, Clemens JD. 2013. 5 year efficacy of a bivalent killed whole-cell oral cholera vaccine in Kolkata, India: a cluster-randomised, double-blind, placebo-controlled trial. Lancet Infect Dis 13:1050–1056. 10.1016/S1473-3099(13)70273-1. [DOI] [PubMed] [Google Scholar]
  • 114.Qadri F, Ali M, Lynch J, Chowdhury F, Khan AI, Wierzba TF, Excler J-L, Saha A, Islam MT, Begum YA, Bhuiyan TR, Khanam F, Chowdhury MI, Khan IA, Kabir A, Riaz BK, Akter A, Khan A, Asaduzzaman M, Kim DR, Siddik AU, Saha NC, Cravioto A, Singh AP, Clemens JD. 2018. Efficacy of a single-dose regimen of inactivated whole-cell oral cholera vaccine: results from 2 years of follow-up of a randomised trial. Lancet Infect Dis 18:666–674. 10.1016/S1473-3099(18)30108-7. [DOI] [PubMed] [Google Scholar]
  • 115.Iyer AS, Bouhenia M, Rumunu J, Abubakar A, Gruninger RJ, Pita J, Lino RL, Deng LL, Wamala JF, Ryan ET, Martin S, Legros D, Lessler J, Sack DA, Luquero FJ, Leung DT, Azman AS. 2016. Immune responses to an oral cholera vaccine in internally displaced persons in South Sudan. Sci Rep 6:35742. 10.1038/srep35742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Saha A, Khan A, Salma U, Jahan N, Bhuiyan TR, Chowdhury F, Khan AI, Khanam F, Muruganandham S, Reddy Kandukuri S, Singh Dhingra M, Clemens JD, Cravioto A, Qadri F. 2016. The oral cholera vaccine Shanchol when stored at elevated temperatures maintains the safety and immunogenicity profile in Bangladeshi participants. Vaccine 34:1551–1558. 10.1016/j.vaccine.2016.02.020. [DOI] [PubMed] [Google Scholar]
  • 117.Baik YO, Choi SK, Olveda RM, Espos RA, Ligsay AD, Montellano MB, Yeam JS, Yang JS, Park JY, Kim DR, Desai SN, Singh AP, Kim IY, Kim CW, Park S. 2015. A randomized, non-inferiority trial comparing two bivalent killed, whole cell, oral cholera vaccines (Euvichol vs Shanchol) in the Philippines. Vaccine 33:6360–6365. 10.1016/j.vaccine.2015.08.075. [DOI] [PubMed] [Google Scholar]
  • 118.Levine MM, Kaper JB. 1995. Live oral cholera vaccine: from principle to product. Bull Inst Pasteur 93:243–253. 10.1016/0020-2452(96)85758-7. [DOI] [Google Scholar]
  • 119.Desai SN, Cravioto A, Sur D, Kanungo S. 2014. Maximizing protection from use of oral cholera vaccines in developing country settings. Hum Vaccin Immunother 10:1457–1465. 10.4161/hv.29199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Saluja T, Mogasale VV, Excler J-L, Kim JH, Mogasale V. 2020. An overview of Vaxchora, a live attenuated oral cholera vaccine. Hum Vaccin Immunother 16:42–50. 10.1080/21645515.2019.1644882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Suharyono, Simanjuntak C, Totosudirjo H, Witham N, Punjabi N, Burr D, Sorenson K, Heppner DG, Losonsky G, Clemens J, Lim YL, Wasserman SS, Kaper J, Levine MM, Rifai AR, Cryz S. 1992. Safety and immunogenicity of single-dose live oral cholera vaccine CVD 103-HgR in 5-9-year-old Indonesian children. Lancet 340:689–694. 10.1016/0140-6736(92)92231-4. [DOI] [PubMed] [Google Scholar]
  • 122.Herzog C. 2016. Successful comeback of the single-dose live oral cholera vaccine CVD 103-HgR. Travel Med Infect Dis 14:373–377. 10.1016/j.tmaid.2016.07.003. [DOI] [PubMed] [Google Scholar]
  • 123.Levine MM. 2010. Immunogenicity and efficacy of oral vaccines in developing countries: lessons from a live cholera vaccine. BMC Biol 8:129. 10.1186/1741-7007-8-129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Holmgren J. 2021. An update on cholera immunity and current and future cholera vaccines. Trop Med Infect Dis 6:64. 10.3390/tropicalmed6020064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Hubbard TP, Billings G, Dörr T, Sit B, Warr AR, Kuehl CJ, Kim M, Delgado F, Mekalanos JJ, Lewnard JA, Waldor MK. 2018. A live vaccine rapidly protects against cholera in an infant rabbit model. Sci Transl Med 10:eaap8423. 10.1126/scitranslmed.aap8423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Ryan ET, Calderwood SB. 2000. Cholera vaccines. Clin Infect Dis 31:561–565. 10.1086/313951. [DOI] [PubMed] [Google Scholar]
  • 127.Levine MM, Chen WH, Kaper JB, Lock M, Danzig L, Gurwith M. 2017. PaxVax CVD 103-HgR single-dose live oral cholera vaccine. Expert Rev Vaccines 16:197–213. 10.1080/14760584.2017.1291348. [DOI] [PubMed] [Google Scholar]
  • 128.Clements JD, Freytag LC. 2016. Parenteral vaccination can be an effective means of inducing protective mucosal responses. Clin Vaccine Immunol 23:438–441. 10.1128/CVI.00214-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Akter A, Kelly M, Charles RC, Harris JB, Calderwood SB, Bhuiyan TR, Biswas R, Xu P, Kováč P, Qadri F, Ryan ET. 2021. Parenteral vaccination with a cholera conjugate vaccine boosts vibriocidal and anti-OSP responses in mice previously immunized with an oral cholera vaccine. Am J Trop Med Hyg 104:2024–2030. 10.4269/ajtmh.20-1511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Hsiao A, Desai SN, Mogasale V, Excler J-L, Digilio L. 2017. Lessons learnt from 12 oral cholera vaccine campaigns in resource-poor settings. Bull World Health Organ 95:303–312. 10.2471/BLT.16.175166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Khan AI, Levin A, Chao DL, DeRoeck D, Dimitrov DT, Khan JAM, Islam MS, Ali M, Islam MT, Sarker AR, Clemens JD, Qadri F. 2018. The impact and cost-effectiveness of controlling cholera through the use of oral cholera vaccines in urban Bangladesh: a disease modeling and economic analysis. PLoS Negl Trop Dis 12:e0006652. 10.1371/journal.pntd.0006652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Karlsson SL, Ax E, Nygren E, Källgård S, Blomquist M, Ekman A, Benktander J, Holmgren J, Lebens M. 2014. Development of stable Vibrio cholerae O1 Hikojima type vaccine strains co-expressing the Inaba and Ogawa lipopolysaccharide antigens. PLoS One 9:e108521. 10.1371/journal.pone.0108521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Chowdhury F, Ali Syed K, Akter A, Rahman Bhuiyan T, Tauheed I, Khaton F, Biswas R, Ferdous J, Al Banna H, Ross AG, Mc Millan N, Sharma T, Kanchan V, Pal Singh A, Gill D, Lebens M, Nordqvist S, Holmgren J, Clemens JD, Qadri F. 2021. A phase I/II study to evaluate safety, tolerability and immunogenicity of Hillchol, an inactivated single Hikojima strain based oral cholera vaccine, in a sequentially age descending population in Bangladesh. Vaccine 39:4450–4457. 10.1016/j.vaccine.2021.06.069. [DOI] [PubMed] [Google Scholar]
  • 134.Ahmed T, Svennerholm A-M, Tarique AA, Sultana GNN, Qadri F. 2009. Enhanced immunogenicity of an oral inactivated cholera vaccine in infants in Bangladesh obtained by zinc supplementation and by temporary withholding breast-feeding. Vaccine 27:1433–1439. 10.1016/j.vaccine.2008.12.036. [DOI] [PubMed] [Google Scholar]
  • 135.Hashim R, Khatib AM, Enwere G, Park JK, Reyburn R, Ali M, Chang NY, Kim DR, Ley B, Thriemer K, Lopez AL, Clemens JD, Deen JL, Shin S, Schaetti C, Hutubessy R, Aguado MT, Kieny MP, Sack D, Obaro S, Shaame AJ, Ali SM, Saleh AA, von Seidlein L, Jiddawi MS. 2012. Safety of the recombinant cholera toxin B subunit, killed whole-cell (rBS-WC) oral cholera vaccine in pregnancy. PLoS Negl Trop Dis 6:e1743. 10.1371/journal.pntd.0001743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Grout L, Martinez-Pino I, Ciglenecki I, Keita S, Diallo AA, Traore B, Delamou D, Toure O, Nicholas S, Rusch B, Staderini N, Serafini M, Grais RF, Luquero FJ. 2015. Pregnancy outcomes after a mass vaccination campaign with an oral cholera vaccine in Guinea: a retrospective cohort study. PLoS Negl Trop Dis 9:e0004274. 10.1371/journal.pntd.0004274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Khan AI, Ali M, Chowdhury F, Saha A, Khan IA, Khan A, Akter A, Asaduzzaman M, Islam MT, Kabir A, You YA, Saha NC, Cravioto A, Clemens JD, Qadri F. 2017. Safety of the oral cholera vaccine in pregnancy: retrospective findings from a subgroup following mass vaccination campaign in Dhaka, Bangladesh. Vaccine 35:1538–1543. 10.1016/j.vaccine.2017.01.080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Ali M, Kim P, Zaman K, Clemens J. 2019. Herd protection of unvaccinated adults by oral cholera vaccines in rural Bangladesh. Int Health 11:229–234. 10.1093/inthealth/ihy085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Clemens JD, Harris JR, Khan MR, Kay BA, Yunus M, Svennerholm A-M, Sack DA, Chakraborty J, Stanton BF, Khan MU, Atkinson W, Holmgren J. 1986. Field trial of oral cholera vaccines in Bangladesh. Lancet ii:124–127. 10.1016/S0140-6736(86)91944-6. [DOI] [PubMed] [Google Scholar]
  • 140.Longini IM, Jr, Nizam A, Ali M, Yunus M, Shenvi N, Clemens JD. 2007. Controlling endemic cholera with oral vaccines. PLoS Med 4:e336. 10.1371/journal.pmed.0040336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Chowdhury FR, Nur Z, Hassan N, von Seidlein L, Dunachie S. 2017. Pandemics, pathogenicity and changing molecular epidemiology of cholera in the era of global warming. Ann Clin Microbiol Antimicrob 16:10. 10.1186/s12941-017-0185-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Chowdhury F, Bhuiyan TR, Akter A, Bhuiyan MS, Khan AI, Tauheed I, Ahmed T, Ferdous J, Dash P, Basher SR, Hakim A, Lynch J, Kim JH, Excler J-L, Kim DR, Clemens JD, Qadri F. 2020. Augmented immune responses to a booster dose of oral cholera vaccine in Bangladeshi children less than 5 years of age: revaccination after an interval of over three years of primary vaccination with a single dose of vaccine. Vaccine 38:1753–1761. 10.1016/j.vaccine.2019.12.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Chowdhury F, Bhuiyan TR, Akter A, Bhuiyan MS, Khan AI, Hossain M, Tauheed I, Ahmed T, Islam S, Rafique TA, Siddique SA, Harun NB, Islam K, Clemens JD, Qadri F. 2020. Immunogenicity of a killed bivalent whole cell oral cholera vaccine in forcibly displaced Myanmar nationals in Cox’s Bazar, Bangladesh. PLoS Negl Trop Dis 14:e0007989. 10.1371/journal.pntd.0007989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.World Health Organization. 2020. Management of the patient with cholera. World Health Organization, Geneva, Switzerland. https://apps.who.int/iris/bitstream/handle/10665/58493/WHO_CDD_SER_91.15_REV.1.pdf. Accessed 4 August 2020. [Google Scholar]
  • 145.World Health Organization. 2002. Reduced osmolarity: oral rehydration salts (ORS) formulation: a report from a meeting of experts jointly organised by UNICEF and WHO: UNICEF house, New York, USA, 18 July 2001. [Google Scholar]

Articles from Clinical Microbiology Reviews are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES