Emerg Infect DisEIDEmerging Infectious Diseases1080-60401080-6059Centers for Disease Control and Prevention17553261273845907-001510.3201/eid1305.070015DispatchChikungunya Virus in US Travelers Returning from India, 2006Chikungunya Virus in US TravelersLanciottiRobert S.*KosoyOlga L.*LavenJaneen J.*PanellaAmanda J.*VelezJason O.*LambertAmy J.*CampbellGrant L.*Centers for Disease Control and Prevention, Fort Collins, Colorado, USAAddress for correspondence: Robert S. Lanciotti, Diagnostic and Reference Laboratory, Arbovirus Diseases Branch, Centers for Disease Control and Prevention, 3150 Rampart Rd, Fort Collins, CO 80521, USA; email: rsl2@cdc.gov52007135764767

Chikungunya virus (CHIKV), a mosquitoborne alphavirus; is endemic in Africa and Asia. In 2005–2006, CHIKV epidemics were reported in islands in the Indian Ocean and in southern India. We present data on laboratory-confirmed CHIKV infections among travelers returning from India to the United States during 2006.

Keywords: Chikungunyaalphaviruslaboratory diagnosisRT-PCRdispatch

Chikungunya virus (CHIKV) is a mosquito-transmitted virus (genus Alphavirus, family Togaviridae) usually associated with acute epidemic polyarthralgia. The virus is serologically and genetically most closely related to o’nyong-nyong, Igbo Ora, and, to a lesser extent, Mayaro and Ross River viruses, all of which are associated with acute polyarthralgia (1).

CHIKV epidemics have been described in Africa, the Middle East, India, and Southeast Asia, and may have caused epidemics in the Caribbean and in the United States during the early 19th century (2). CHIKV epidemics can be explosive with large numbers of human cases and rapid virus dissemination. In the Réunion Island epidemic from April 2005 to June 2006, ≈270,000 cases were reported, representing nearly 40% of the population (3). Aedes aegypti is the principal vector; however, in recent epidemics in Réunion Island and southern India, Ae. albopictus has been co-implicated (4,5). In Africa, CHIKV is maintained in an enzootic cycle involving primates, but in Asia and in recent large epidemics, the human-mosquito cycle predominates, possibly including mechanical transmission (6). Symptoms are characterized by acute onset of joint pain, followed by myalgia, fever, and rash with recovery usually within weeks.

Laboratory diagnosis of CHIKV infection is accomplished by serologic methods, virus isolation, and reverse transcription–PCR (RT-PCR). A typical serologic algorithm involves testing acute- and convalescent-phase serum specimens for immunoglobulin M (IgM) and IgG antibody, followed by a plaque reduction neutralization test (PRNT). Virus isolation and RT-PCR are normally used with early acute-phase specimens (before day 5 post-onset) because duration of viremia is typically 2–4 days.

Recent CHIKV outbreaks have been reported in several islands in the Indian Ocean as well as in southern India, where >1 million cases were reported in 2006 (4,7). CHIKV infections have also been documented in travelers returning from these areas (3,7). We report confirmed CHIKV infections among 35 travelers returning from overseas travel; 33 were returning from India and 2 from Réunion Island (Table 1).

Diagnostic test results for 35 travelers infected with chikungunya virus (CHIKV), 2006*
SampleIgM ELISA†IgG ELISA†PRNT‡Virus isolation
(Vero cells)RT-PCR§Viremia, PFU/mL¶Days from onset of illness
to collectionState of US
residenceReturn date, 2006
1
17.7
3.2
640


NA
0
NJ
10/12
2
1.7
1.7
<10

+
104.0
1
CA
Before 11/28
3
1.2
1.1
ND
+
+
104.1
1
IL
9/29
4
1.8
NS
ND
+
+
106.8
2
CA
Before 9/16
5
1.2
0.76
ND
+
+
105.1
2
MA
9/10
6
1.8
1.6
<10
+
+
106.0
3
PA
Before 8/20
7
1.6
1.2
ND
+
+
105.3
3
CA
10/2
8
NS
1.4
ND

+
103.9
4
WI
10/9
9
22.0
4.3
5,120


NA
4
CA
Before 10/6
10
1.1
0.95
<10

+
104.5
6
CA
Before 8/13
11
7.4
0.96
40


NA
7
CA
Before 9/23
12
15.0
0.60
320


NA
8
CT
Before 7/6
13
26.2
1.2
160
ND

NA
8
DC
Before 10/16
14
12.9
5.8
1,80
ND

NA
10
CA
Before 9/22
15
38.8
2.3
2,560
ND
ND
NA
19
CT
Before 7/6
16
12.7
1.5
640
ND

NA
20
IL
8/23
17
16.9
4.8
640
ND

NA
30
IL
6/25
18
8.3
3.7
640
ND
ND
NA
31
HI
Before 8/2
19
6.6
1.5
640
ND

NA
31
CA
Before 8/13
20
2.6
6.8
320
ND

NA
34
MD
Before 3/20
21
5.0
NS
640
ND

NA
34
IL
Before 9/22
22
15.1
4.1
1,280
ND

NA
38
CA
Before 10/2
23
4.3
5.9
2,560
ND

NA
42
PA
Before 8/20
24
5.6
NS
320
ND

NA
43
Unknown
Unknown
25
3.1
3.7
640
ND

NA
44
IL
Before 9/12
26
26.6
13.4
5,120
ND
ND
NA
48
SC
6/24
27
30.7
6.6
5,120
ND

NA
61
CA
Before 10/26
28
6.1
11.4
2,560
ND

NA
62
CT
Before 10/3
29
9.4
16.0
640
ND

NA
63
CA
8/23
30
7.5
8.4
1,280
ND

NA
71
MN
6/8
31
5.3
5.8
640
ND

NA
71
MN
6/8
32
3.7
16.1
320
ND

NA
75
LA
Before 3/30
33
9.8
10.1
2,560
ND

NA
92
IL
7/9
34
7.5
3.1
160
ND

NA
101
PA
Before 10/13
3524.611.920,480NDNAUnknownILBefore 11/08

*IgM, immunoglobulin M; PRNT, plaque reduction neutralization test; RT-PCR, reverse transcription–PCR; NA, not applicable; ND, not done
(sampled depleted); NS, nonspecific reaction in ELISA.
†Values are patient sample optical densities divided by a negative control optical density; values >3 are positive.
‡Values are 90% plaque reduction neutralization titers.
§Real-time, fluorescence-based assay for detecting CHIKV RNA; positive samples had crossing threshold values <37 with both primer sets.
¶Estimated CHIKV PFU/mL by real-time RT-PCR using a standard curve generated with plaque-titrated/calibrated CHIKV standards.

The Study

Serum samples were received by the Centers for Disease Control and Prevention (Fort Collins, CO, USA) from April 2006 to December 2006 as part of routine diagnostic and reference services available to public health laboratories. A total of 106 serum samples were received from persons returning from regions with epidemics or where CHIKV is endemic (79 from India and the Indian Ocean islands and 27 from Africa) with compatible CHIKV illness and submitted by state public health laboratories. Serum samples were tested for antibodies to several viruses known to occur in the region of travel and residence by IgM capture ELISA and a standard IgG ELISA (8,9). The 35 CHIKV IgM- and IgG-positive specimens were tested by using a PRNT (90% reduction cutoff) with several related alphaviruses (Sindbis, o’nyong-nyong, and Semliki Forest viruses) to confirm specificity of reactivity (10). A ≥4-fold neutralizing titer difference between antibody to CHIKV and antibodies to other alphaviruses indicated a CHIKV-specific antibody response. IgM-positive and PRNT specificity–confirmed specimens were classified as recent CHIKV infections (Table 1).

All serum specimens were tested by a quantitative, real-time, fluorescent probed–based RT-PCR assay for CHIKV RNA. Two primer probe sets were designed in unique regions of the viral genome and reacted specifically with CHIKV RNA and not with related or unrelated viruses (Table 2). Both sets showed an analytical sensitivity <1 PFU, and CHIKV was detected in virus-spiked serum samples at a concentration of 10 PFU/mL (75 μL of serum assayed). Eight serum specimens showed positive results by the real-time assay; all were acute-phase specimens with number of days post-onset of illness reported as <6. Viral titers of these specimens were estimated by quantitative RT-PCR that used CHIKV quantity standards (determined by plaque assay) to generate a standard curve. Titers of 8 specimens ranged from 103.9 PFU/mL to 106.8 PFU/mL.

Sensitivity and specificity of chikungunya virus (CHIKV) oligonucleotide primers used in real-time reverse transcription–PCR assay
PrimerGenome position*Sequence (5′→3′)Sensitivity†Specificity‡
CHIKV 874
874–894
AAAGGGCAAACTCAGCTTCAC


CHIKV 961
961–942
GCCTGGGCTCATCGTTATTC
0.3
CHIKV
CHIKV 899-FAM§
899–923
CGCTGTGATACAGTGGTTTCGTGTG


CHIKV 6856
6856–6879
TCACTCCCTGTTGGACTTGATAGA


CHIKV 6981
6981–6956
TTGACGAACAGAGTTAGGAACATACC
0.9
CHIKV
CHIKV 6919-FAM6919–6941AGGTACGCGCTTCAAGTTCGGCG

*On the basis of CHIKV prototype strain S27, GenBank accession no. NC_004162.
†Absolute no. of PFU detected in triplicate testing.
‡No reactivity was observed with the following viruses: o’nyong nyong, Ross River, Mayaro, Semliki Forest, Sindbis, western
equine encephalitis, eastern equine encephalitis, and Venezuelan equine encephalitis subtypes 1AB, 1C, 1D, and 1E.
§Primer labeled at the 5′ terminus with 5-FAM and 3′ Black Hole Quencher 1 (Operon Biotechnologies Inc., Huntsville, AL, USA).

All acute-phase specimens (on or before day 8 postonset) were also tested for CHIKV by virus isolation in Vero cells. Isolation was performed by using a recently developed protocol in which cells were grown in glass shell vials and centrifuged to enhance viral infectivity (J.O. Velez, unpub. data). Five serum specimens displayed prominent and characteristic cytopathic effect on day 2 postinfection, and virus was identified as CHIKV by RT-PCR. All virus isolates were obtained from acute-phase specimens that also were positive by RT-PCR. Three serum specimens (samples 2, 8, and 10) showed positive RT-PCR results, but CHIKV was not isolated from these specimens. In these 3 specimens, inability to isolate virus may have been related to viral titers, which were lower than most of the virus isolation–positive samples, or to handling or storage of these samples. All 8 virus-positive specimens (whether positive by RT-PCR, virus isolation, or both) were collected <7 days postonset and were negative for IgM and IgG antibodies to CHIKV. Nearly all of the specimens collected <7 days postonset were positive by 1 of the virus-based tests. The 2 exceptions, samples 1 and 9, were positive for IgM and IgG antibodies to CHIKV and had high PRNT titers. These findings indicate that these samples were not true acute-phase specimens; the true onset or collection date had likely been reported incorrectly.

To identify the strain of CHIKV in these specimens, a 2,122-bp fragment from the structural region of the genome (nucleotide positions 9,648–11,770) was amplified from all 8 virus-positive specimens by RT-PCR and subjected to nucleic acid sequencing with previously described primers (11). All 8 sequences showed nucleotide identity >99.7% (GenBank accession nos. EF451142–EF451149). BLAST analysis (www.ncbi.nlm.nih.gov/blast) of the 8 sequences showed that the highest percentage identity was to CHIKV strains recently isolated from travelers returning from Indian Ocean islands (Réunion, Mauritius, and Seychelles). Percentage identity matches between the 8 viruses and Indian Ocean CHIKV strains were >99.5%, with 5 to 8 mismatches occurring randomly. In comparison, percentage identities of the 8 viruses to CHIKV prototype S27 or to a strain previously isolated from India (Nagpur/653496) were 95.1% and 94.4%, respectively.

Conclusions

The data reported confirm that the widespread CHIKV epidemic in southern India has infected US travelers. CHIKV infections among international travelers are not unexpected; in 2005–2006, ≈800 CHIKV infections were reported in France, primarily in travelers returning from Réunion Island (3). The more noteworthy observation of this study with potential public health ramifications is that high levels of infectious virus were detected in returning travelers. Primary vectors for CHIKV are Ae. aegypti and Ae. albopictus, which are established in several southeastern coastal states in the United States. Vector competence studies of Ae. aegypti and Ae. albopictus strains from the United States, as well as strains from the Caribbean and South America, showed that a titer of ≈104 PFU/mL in monkeys resulted in productive infection with virus dissemination in these mosquitoes, with Ae. albopictus showing higher infection and dissemination rates (12). The level of viremia reported in most of these imported CHIKV infections, >104 PFU/mL, could be sufficient to infect North American vectors, given the appropriate environmental conditions. However, the time of year and place of residence of the returning travelers in this study were not conducive to transmission; only 2 (patients 26 and 32) returned to US regions (South Carolina and Louisiana) known to have populations of Ae. albopictus.

Nevertheless, returning travelers with high viremia levels, who live in areas with established Ae. aegypti and Ae. albopictus populations, could facilitate local transmission in the United States. Clinicians should therefore obtain travel histories from persons with CHIKV-compatible illness and include CHIKV in differential diagnoses when appropriate. Public health laboratories must carefully monitor CHIKV infections of returning travelers and conduct surveillance for CHIKV-infected vectors in high-risk areas to prevent local establishment of a new emerging virus. Diagnostic laboratory personnel involved in virus isolation protocols must be aware of the potential of isolating CHIKV (a biosafety level 3 agent) from patients returning from regions endemic for CHIKV or regions with epidemics and take appropriate safety precautions.

Suggested citation for this article: Lanciotti RS, Kosoy OL, Laven JJ, Panella AJ, Velez JO, Lambert AJ, et al. Chikungunya virus in US travelers returning from India, 2006. Emerg Infect Dis [serial on the Internet]. 2007 May [date cited]. Available from http://www.cdc.gov/eid/content/13/5/764.htm

Dr Lanciotti is chief of the Diagnostic and Reference Laboratory in the Arbovirus Diseases Branch at the Centers for Disease Control and Prevention, Fort Collins, Colorado. His primary research interests are laboratory diagnosis of arbovirus infections and diagnostic test development and support for public health laboratories worldwide.

ReferencesPowers AM, Brault AC, Shirako Y, Strauss EG, Kang W, Strauss JH, Evolutionary relationships and systematics of the alphaviruses.J Virol 2001;75:1011831 10.1128/JVI.75.21.10118-10131.200111581380Carey DE Chikungunya and dengue: a case of mistaken identity.J Hist Med Allied Sci 1971;26:24362 10.1093/jhmas/XXVI.3.2434938938Krastinova E, Quatresous I, Tarantola A. Imported cases of chikungunya in metropolitan France: update to June 2006. Eurosurveillance. 2006;11:E060824.1.Outbreak news. Chikungunya, India.Wkly Epidemiol Rec 2006;81:4091017068865Reiter P, Fontenille D, Paupy C Aedes albopictus as an epidemic vector of chikungunya virus: another emerging problem?Lancet Infect Dis 2006;6:4634 10.1016/S1473-3099(06)70531-X16870524Rao TR, Paul SD, Singh KR Experimental studies on the mechanical transmission of Chikungunya virus by Aedes aegypti.Mosq News 1968;28:4068Centers for Disease Control and Prevention Chikungunya fever diagnosed among international travelers—United States, 2005–2006.MMWR Morb Mortal Wkly Rep 2006;55:1040217008866Martin DA, Muth DA, Brown T, Johnson AJ, Karabatsos N, Roehrig JT Standardization of immunoglobulin M capture enzyme-linked immunosorbent assays for routine diagnosis of arboviral infections.J Clin Microbiol 2000;38:1823610790107Johnson AJ, Martin DA, Karabatsos N, Roehrig JT Detection of anti-arboviral immunoglobulin G by using a monoclonal antibody-based capture enzyme-linked immunosorbent assay.J Clin Microbiol 2000;38:18273110790108Lindsey HS, Calisher CH, Mathews JH Serum dilution neutralization test for California group virus identification and serology.J Clin Microbiol 1976;4:503101002829Schuffenecker I, Iteman I, Michault A, Murri S, Frangeul L, Vaney M, Genome microevolution of chikungunya viruses causing the Indian Ocean outbreak.PLoS Med 2006;3:105870 10.1371/journal.pmed.0030263Turell MJ, Beaman JR, Tammariello RF Susceptibility of selected strains of Aedes aegypti and Aedes albopictus (Diptera: Culicidae) to chikungunya virus.J Med Entomol 1992;29:49531313111