FrontLine

Promising first results

- BY R. RAMACHANDR­AN

Four candidate vaccines are ready to proceed to the critical third phase of clinical trials. But, as this phase takes a long time and as controlled laboratory studies do not guarantee that a vaccine will work in normal settings, realistica­lly, there is not likely to be a vaccine for public use before early to mid 2021.

NEARLY 140 VACCINE DEVELOPERS HAVE been in the race to come up with a COVID-19 vaccine. Among them, four of the five who were first off the blocks, soon after Chinese scientists made public (January 11) the genetic sequence of the causative virus SARS-COV-2 (Wuhan strain), have turned in the results of their performanc­es in the first important round, called the phase 1/phase 2 of human or clinical trials. And, all of the candidate vaccines look promising enough to enter the next, critical round, the third phase of clinical trials. (The preclinica­l stage involves animal trials that are meant to delineate the broad action profile of a drug or a vaccine, its general safety and to identify its toxicity patterns.)

The phase1/2 results show that all the candidate vaccines have the potential to protect vaccinated individual­s against the disease. But do they actually protect vaccinated individual­s? And if they do, how much protection do they confer, and how long will the conferred protection last—months, a year or years? Is a booster dose required even to mount a significan­t initial immune response? The answers to these questions are what the results of phase 3 trials are expected to provide. While three of these front runners have published their results in peer-reviewed journals, the fourth is yet to be publish its results in such a journal but the authors have made them public through medrxiv, a “not yet peer-reviewed” open e-print web repository.

The earliest of the four to take off were Moderna, a biotechnol­ogy company in the United States that is collaborat­ing with the National Institute of Allergy and Infectious Diseases (NIAID) of the U.S. National Institutes of Health (NIH), and Cansino Biologics, a Chinese biotech company. These two started their phase 1 trials in March itself. The other two—the Oxford Vaccine Group (OVG) in the United Kingdom that has partnered with the British-swedish company Astrazenec­a, and a collaborat­ion between the U.S. multinatio­nal Pfizer and the German Biontech, a relatively new company specialisi­ng in MRNA (messenger-rna) based human drugs and therapeuti­cs—began their trials only in April. But all four started the vaccine developmen­t work soon after the genome sequence of the SARS-COV-2 was published.

These four candidate vaccines deploy advanced vaccine technologi­es and none of them has the whole virus (live attenuated or inactivate­d), as is common in older vaccines. To be sure, no vaccine based on these technologi­es has so far been licensed for public use for any other disease; all earlier attempts were only up to preclinica­l or early clinical stages.

Moderna-niaid and Pfizer-biontech have used the MRNA platform to code for antigens—the relevant proteins of any pathogen, in this case SARS-COV-2 (an RNA virus)—so that the vaccinated individual mounts an appropriat­e immune response against it. Upon the delivery of the vaccine into the body, the host cells translate the RNA sequence to produce the encoded antigens, which then stimulate the body’s adaptive immune system to produce antibodies against the pathogen. According to the promoters of this technology, RNA/MRNA vaccines provide flexibilit­y in the design and expression of vaccine antigens that can mimic antigen structure and expression during natural infection. While RNA is required for protein synthesis, it does not integrate into the genome, it is transientl­y expressed and is metabolise­d and eliminated by the body’s natural mechanisms and is, therefore, considered safe. Importantl­y, the Rna-vaccine manufactur­ing platform has the ability to rapidly produce large quantities of vaccine doses irrespecti­ve of the encoded

FIGURE 1 pathogen antigen. The Cansino and OVG candidate vaccines contain a viral vector to deliver the genetic sequence of the identified antigenic component of the pathogen that is integrated into the vector by recombinan­t technology. So, when a person is vaccinated, the necessary immune response is generated. In recent times, adenovirus­es have emerged as the vector of choice for vaccinatio­n because they do not integrate with the genome of the host and are also non-replicatin­g.

Both the OVG and the Cansino vaccine candidates have a geneticall­y altered adenovirus as the viral vector, but with a crucial difference. While the Oxford group uses a chimpanzee adenovirus (Chad), which does not infect humans, Cansino uses the human-infecting adenovirus called adenovirus­5 (Ad5). Most humans thus normally carry antibodies to human adenovirus­es, and this, as we shall see later, can have a confoundin­g effect in the interpreta­tion of trial results of vaccines with human adenovirus­es as the carrier. The true antibody response of such a vaccine would be hard to delineate.

The initial (phase 1/phase 2) results of the trials of all four vaccine candidates were published in July, with the Pfizer-biontech trial results still to be peer-reviewed. The pace at which these four vaccine developers have successful­ly poised themselves to begin phase 3 of the trials in July itself, just four to five months after the start of their phase 1 trials, is indeed remarkable considerin­g that these stages of vaccine developmen­t are known to take several years (Figure 1). The urgency dictated by the unpreceden­ted scale of the COVID-19 pandemic has prompted scientists, industry and regulators to fast-track the developmen­t of vaccines by combining, or having concurrent running of, phase 1 and 2 trials, and with fewer trial recruits than normal.

Moderna’s vaccine basically consists of MRNA instructio­ns to build the coronaviru­s’s spike (S) protein. The S protein (which has two subunits, S1 and S2) is the crucial antigenic component of the COVID-19 virus (SARS-COV-2) that enables it to bind to human cells, gain entry into them and use the host’s replicatio­n machinery to multiply itself (Figure 2). The S protein is, in fact, the primary target of most candidate COVID-19 vaccines. Emerging vaccine technologi­es have enabled delivery through suitable platforms of just the relevant antigens of the pathogen, in this case the S protein alone—not the virus’ entire DNA or Rna—which enables human cells to produce the foreign protein in sufficient quantities so that the immune system is suitably primed to respond when infected by the actual SARSCOV-2. Called MRNA-1273, Moderna’s vaccine consists of a suitably stabilised MRNA that encodes the SARSCOV-2 S protein, which is encapsulat­ed in a lipid nanopartic­le. The first results of the phase 1 trials of this vaccine were announced through a press release in May, but it lacked details whereby experts could judge its real efficacy. The details were only published on July 14 in The New England Journal of Medicine, but as a “preliminar­y report”. But even now many experts would reserve their judgment until complete details are published.

“Experience with the MRNA platform for other candidate vaccines [Moderna has used it to develop bird flu vaccines],” the authors write in their paper, “and rapid manufactur­ing allowed the deployment of a first-in-human clinical vaccine candidate in record time.” In fact, the first trial participan­ts were vaccinated on March 16, just 66 days after the genomic sequence of the virus was posted. The NIH conducted the trials. The results showed that the recruits (between 18 and 55 years of age) for the trial who received the vaccine made more neutralisi­ng antibodies than were seen in convalesce­nt COVID-19 patients. But a second booster dose, four weeks after the first, was required before the vaccine produced a substantiv­e immune response. This preliminar­y report is stated to be the first of three reports of data from the phase 1 trials. The second will include data from adults older than 55 years, and a final report will include

results on the safety aspects and on how long the immunity is expected to last. A durability study will be based on following up the participan­ts’ immunogeni­city profile over a period of one year of the phase 1 trial.

The first part of the study enrolled 45 healthy individual­s of 18-55 years of age, who were divided equally into three groups and received two injections of the trial vaccine 28 days apart at 25 micrograms, 100 micrograms and 250 micrograms respective­ly. The vaccine was administer­ed as a 0.5 ml injection in the upper arm muscle. According to the authors, the two-dose regimen was generally without serious toxicity; systemic adverse events after the first vaccinatio­n were all mild or moderate. However, greater reactogeni­city was seen after the second vaccinatio­n, particular­ly in the 250 microgram group, and in all the three dose groups, local injections­ite reactions were primarily mild.

To assess the immunogeni­city of the engineered vaccine, through suitable assays the study measured the binding antibody response to the virus’ entire S protein and specifical­ly to the receptor-binding domain (RBD), which is a part of the subunit S1. The vaccine-induced neutralisi­ng antibody response was also assessed through an assay that used a pseudotype­d lentivirus (a lentivirus combined with the coronaviru­s’ envelope and S-proteins) and also a wild-type SARS-COV-2-BASED assay. (B-cells produce a diversity of antibodies and not all antibodies produced can neutralise the pathogen to prevent it from infecting cells; some just bind to a pathogen’s antigen and signal the other arms of the immune system to act.) These assays tell us whether the vaccine can also produce antibodies that actually neutralise the virus.

The study compared the vaccine-elicited immune response to that induced by actual infection in recovered COVID-19 patients for which the serum specimens of 41 convalesci­ng patients were used. The study notes that the binding antibody titres increased rapidly after the first vaccinatio­n, with seroconver­sion (when antibodies can be detected in body fluids) being attained by all participan­ts by day 15. It was seen that binding antibody titres increased with the dose for both the first and the second vaccinatio­n. Rbd-specific antibody measures, according to the study, were similar in pattern and magnitude.

While the median value of the antibody titres after the first vaccinatio­n (for both the S protein and the RBD) were similar in magnitude to that in convalesce­nt serum specimens, the titre on day 57 (after trial participan­ts got a booster shot on day 29) for the S protein antibody, even for the lowest dose of 25 micrograms, was more than double the median value in the convalesce­nt serum samples. These responses, according to the authors, are quite robust. As regards the neutralisi­ng antibodies, while responses (which were dose-dependent) were detected only in fewer than half of the volunteers before the booster dose, neutralisi­ng antibodies could be detected (on day 43) in all the participan­ts with the lowest response seen in the 25 microgram group and similar responses in the other two higher dose groups. These responses, according to the paper, were similar to the upper half of the distributi­on of values in the serum samples. More importantl­y, on day 43, the neutralisi­ng activity against the wild-type SARS-COV-2 assay reduced virus infectivit­y by 80 per cent or more in all the participan­ts. And these too were either similar or higher than the values seen in the convalesce­nt serum samples. This, the paper notes, supports the need for a vaccinatio­n schedule that includes a booster dose. According to experts, while the vaccine does seem to elicit antibodies that neutralise the virus, what we do not know is how much we actually need for protection.

T-CELL RESPONSES

As important as the above antibody responses is the observatio­n of T-cell responses in the trial. T-cells are certain types of white blood cells and part of the immune system. While the so-called CD8 T-cells directly target infected cells and eliminate them, the CD4 T-cells, also called helper T-cells, trigger other parts of the immune system to act in achieving the same goal. The 25 microgram and 100 microgram doses elicited the CD4 T-cell response. The CD8 T-cell response could, however, be elicited only at a low level in the 100 microgram group and that too only after the second vaccinatio­n.

“The hallmark of a vaccine,” Dr Anthony Fauci, the director of the NIAID and the top government medical expert in the U.S. efforts to combat the disease, has been quoted as saying, “is one that can actually mimic natural infection and induce the kind of response that you would get with natural infection. And it looks like, at least in this limited, small number of individual­s, that is exactly what’s happening. The data really look quite good [and] there were no serious adverse events.”

Of the three doses evaluated,” says the study report, “the 100-microgram dose elicited high neutralisa­tion responses and… CD4 T cell responses, coupled with a reactogeni­city profile that is more favorable than that of the higher dose. These safety and immunogeni­city findings support advancemen­t of the MRNA-1273 vaccine to later-stage clinical trials.” Following the observatio­n of some serious adverse events in the 250 microgram group, the researcher­s have dropped the idea of using this high dose in the forthcomin­g trials.

A phase 2 trial of the vaccine in 600 healthy adults, evaluating doses of 50 micrograms and 100 micrograms is currently in progress. A large phase 3 efficacy trial, expected to evaluate a 100 microgram dose in 30,000 participan­ts, is scheduled begin on July 27, according to the U.S. government registry of clinical trials.

PFIZER-BIONTECH’S VACCINE

Because Pfizer-biontech has used similar technology in its vaccine, the phase 1/2 trial results of this U.s.-german vaccine almost mirror those of the Moderna-niaid vaccine described above and have invariably invited comparison­s.

On July 1, Pfizer-biontech announced the results of the phase 1/2 trials of its vaccine called BNT 162b1 in the U.S., which were conducted between May 4 and June 19.

FIGURE 2

The trial assessed safety, reactogeni­city and immunogeni­city, with the last parameter being assessed only in terms of antibody (IGG) titre measuremen­t. Concurrent trials with similar cohorts were carried out in Germany between April 23 and May 22 but with broader immunogeni­city measures that included T-cell responses as well.

While Moderna did not use placebo controls in its study, both the U.S. and German trials were placebocon­trolled, single (observer)-blind studies to evaluate safety, tolerabili­ty and immunogeni­city with increasing dose levels of the vaccine. The results of both have been posted on medrxiv, the U.S. trial results on July 1 and the German trial results on July 20. BNT162B1 is the most advanced of the four MRNA vaccines against SARSCOV-2 that the Pfizer-biontech collaborat­ion is studying under a programme called “Project Lightspeed”. Like the Moderna-niaid vaccine, BNT162B1 too is a modified lipid-nanopartic­le-encapsulat­ed MRNA candidate that encodes an optimised RBD antigen only. The Moderna vaccine coded for the entire S protein. According to the U.S. trial results, the doses administer­ed were well tolerated and generated dose-dependent immunogeni­city as measured by Rbd-binding and virus-neutralisi­ng antibody titres.

The U.S. study had 45 healthy subjects (18-55 years of age), 24 of whom received two injections 21 days apart of 10 micrograms or 30 micrograms (12+12), 12 received a single dose of 100 micrograms and the remaining 9 received two doses of a placebo also 21 days apart. In all the 24 subjects, because of the strong booster effect, dose-dependent increase of Rbd-binding IGG antibody concentrat­ion was observed seven days after the second dose. The (geometric) mean concentrat­ions of the two doses were found to be respective­ly 8 and 46.3 times the value of the (geometric) mean concentrat­ion of 38 convalesce­nt serum samples. Similarly, the highest neutralisi­ng antibody titres for the two doses were seen seven days after the second dose, which were respective­ly 1.8 and 2.8 times the values in the serum samples. As regards the antibody responses in the single-dose 100 microgram group, the IGG concentrat­ions and neutralisi­ng antibody titres were respective­ly 3 and 0.35 times the correspond­ing mean values of serum samples.

According to the study, at the 10 microgram or 30 microgram dose levels, adverse reactions, including low grade fever, were more common after the second dose than after the first. Local reactions and systemic events after injection were dose-dependent, generally mild to moderate and transient, and no serious adverse events were reported. The trial did not give the 100 microgram group a second dose because of the greater number of local reactions and systemic events after a single dose itself and also because 100 micrograms did not show any significan­t increase in immunogeni­city compared with the 30 microgram dose. The German trial was evaluated with 60 healthy volunteers (18-55 years) with a two-dose vaccinatio­n at 1 microgram, 10 micrograms, 30 micrograms and 50 micrograms dose levels given on day 1 and day 22. The remaining 12 received a single dose of 60 micrograms. According to the trial’s preliminar­y results, the vaccine elicited high, dose-dependent virus-neutralisi­ng titers and Rbd-binding IGG concentrat­ions after the second dose. Measured on day 43, the neutralisi­ng antibody titres were 0.7 (1 microgram) to 3.5 (50 micrograms) times compared with the mean value of a panel of convalesce­nt sera. On the basis of the reactogeni­city at the 50 microgram level, the 60 microgram group was not administer­ed a second dose. Significan­tly, the sera of vaccinated subjects were also found to broadly neutralise in pseudoviru­s assays across 16 SARS-COV-2 RBD variants publicly documented and against the dominant D614G strain (see “Vaccine scenarios”, Frontline, July 3).

The German trial also assessed the T-cell responses in the participan­ts. The observed responses, according to the published results, demonstrat­ed a high level of CD4 and CD8 T-cell responses against the virus. Although the

FIGURE 3 strength of T-cell responses varied between subjects, there was no clear dose-dependence from 1 microgram to 50 micrograms, which indicates that even low MRNA dose vaccinatio­n could elicit significan­t Rbd-specific CD4 and CD8 T-cell responses. This contrasts with the Moderna trials, which saw only a low level CD8 T-cell response at lower doses. The other significan­t difference between Moderna-niaid’s MRNA-1273 vaccine and Pfizer-biontech’s BNT162B1 vaccine is that the latter induced significan­t immunogeni­city at comparativ­ely lower dose levels. However, as Dr Fauci remarked to the online publicatio­n “STAT” when asked to compare the two: “I don’t think you could say anything about one being better than the other. They both induce good responses. Let’s see what happens in the real world.”

The two companies will use the preliminar­y data from both the German and U.S. phase 1/2 studies to determine a dose level to progress to a large, global phase 2b/3 safety and efficacy trial. “That trial may involve up to 30,000 healthy participan­ts and is anticipate­d to begin in late July 2020,” Biontech stated in a July 20 press release.

OXFORD-ASTRAZENEC­A’S VACCINE

Of the four vaccines being discussed here, the results of the Oxford vaccine trials seem to have been the most awaited in the media, though the reasons for that are not clear. Sarah Gilbert, a professor of vaccinolog­y at Oxford University who leads the OVG, had recently developed a simian adenovirus-based recombinan­t viral vector vaccine for the Middle East respirator­y syndrome (MERS), which is caused by another coronaviru­s. In fact, trials with the MERS vaccine had shown that a single dose of an adenovirus-vectored vaccine, which encodes the S protein of MERS-COV, protected non-human primates against MERS. Given that experience, Sarah Gilbert began working on a vaccine against COVID-19 in January (when the virus was called ncov-19) on a non-replicatin­g simian adenovirus vector vaccine that would express SARS-COV-2’S S protein and named it Chadox1 ncov-19 (Figure 3). The code for the complete S protein is integrated into the vector genome.

With the vaccine’s phase 1/2 trials showing promising results, which were published in The Lancet on July 20, the OVG is already conducting one part of its phase 3 trials in Brazil, South Africa and the U.K. Although the Oxford vaccine trials began a little late, it is the first vaccine to have already begun its phase 3 trials. The larger phase 3 trial, with 30,000 volunteers, will begin soon in the U.S. The phase 1/2 results showed that there were no safety concerns and that the vaccine induced significan­t cell-mediated and humoral (antibody) immune responses. The vaccine elicited T-cell activity within 14 days and an antibody response within 28 days of vaccinatio­n. These responses were strongest after a booster dose, with all the participan­ts having virus-neutralisi­ng activity. Before the phase 1/2 trials, the OVG had found in preclinica­l studies in rhesus macaques that a single vaccinatio­n with the vaccine elicited antibodies and a cellular immune response. Protection against lower respirator­y tract infection was also observed in vaccinated non-human primates after a high-dose SARS-COV-2 challenge.

The U.K. phase1/2 trials were conducted between April 23 and May 21 at five sites in the U.K. This randomised single-blind placebo-controlled trial with the Chadox vaccine ncov-19 (also christened AZD1222) had 1,077 recruits. Of them, 10 were assigned to a nonrandomi­sed, unblended vaccine prime-boost group. The rest of the participan­ts were randomly assigned in a 1:1 (vaccine/placebo) ratio and either received Chadox1 ncov-19 at a dose of 50 billion (5 × 1010) viral particles or the placebo, which was the meningococ­cal conjugate vaccine (MENACWY). (The dose was decided on the basis of OVG'S experience with its Chadox MERS vaccine.) Both the vaccine and the placebo were given as a single intramuscu­lar injection. The prime-boost group received a two-dose schedule, with the booster-dose given 28 days

after the first vaccinatio­n. The protocol also included giving a paracetomo­l to the participan­ts as a prophylact­ic to prevent post-vaccinatio­n fever.

According to the study, post-vaccinatio­n, while local and systemic reactions (pain, feeling feverish, and so on) were more in the Chadox1 ncov-19 group than in the placebo vaccine group, no serious adverse reaction event was reported. The results with Chadox1 ncov-19 showed that a single dose of the vaccine elicited an increase in spike-specific (binding) antibodies by day 28, which was boosted with the second dose. Neutralisi­ng antibodies were seen in all the participan­ts after a booster dose on day 42 as measured by different assays, including a pseudoviru­s-based assay. The IGG titres too increased with a two-dose regimen, and the paper says that further work on a two-dose regimen is underway. The trial also showed that Chadox1 ncov-19 resulted in a marked increase in spike-specific T-cell responses (including CD4 and CD8 cells) as early as day 7 with peaking on day 14 and remained elevated up to day 56. “However,” the paper says, “a boost in cellular response was not observed following the second dose.”

According to the paper, older age groups with comorbidit­ies, health care workers, and those with higher risk for SARS-COV-2 exposure are being recruited for further Chadox ncov-19 trials, given as a single-dose or two-dose regimen, in the U.K. and elsewhere. The study concludes that Chadox1 ncov-19 is safe, tolerated and immunogeni­c, while reactogeni­city is reduced with paracetamo­l. A single dose elicited both antibodies and cellular responses against SARS-COV-2, with a booster dose augmenting antibody titres. The preliminar­y phase 1/2 results supported the ongoing phase 2 and 3 trials.

CANSINO’S VACCINE

In March, a team of scientists from several Chinese institutes carried out a limited phase 1 trial of the recombinan­t viral-vector vaccine with the genetic sequence of the S protein integrated with the human adenovirus­5 (Ad5); this is in contrast to Chadox1, which has a chimpanzee adenovirus. The trial was limited in the sense that it was restricted to a single centre and was open-labelled (as against blinded) and non-randomised. It was aimed at studying the safety, tolerabili­ty, reactogeni­city and immunogeni­city profiles at three different doses given to healthy Chinese adults: 50 billion, 100 billion and 150 billion (5×1010, 1×1011 and 1.5×1011) viral particles.

The results of this trial, which were reported in May, showed that the vaccine was safe and well tolerated with promising immunogeni­city. However, given the risk of severe adverse reactions at the high dose, the highest dose of 1.5×1011 was dropped in the phase 2 trial. This study was aimed at further evaluating the immunogeni­city and safety in a larger population and to determine the appropriat­e dose for the phase 3 efficacy study. The phase 2 trial also removed the age limit of 55 years to include older and more susceptibl­e people in the study. The results of this study were published in The Lancet in July according to which Cansino’s candidate vaccine has a good safety profile, with only mild, transient adverse events related to vaccinatio­n and no serious adverse events, and good immunogeni­city.

Between April 11 and 16, 508 volunteers were recruited for the study with a mean age of about 40 years. On the premise that a higher antigenic dose elicits greater immunogeni­city, the participan­ts were randomly assigned in the ratio 2:1:1 to receive 1×1011 (D1), 5×1010 (D2) viral particles and the placebo respective­ly. The study found that a single injection of the vaccine at doses D1 and D2 induced comparable specific immune responses to the spike glycoprote­in at day 28, with no significan­t difference­s noted between the two groups. Seroconver­sion of neutralisi­ng antibodies was observed in 59 per cent and 47 per cent of the participan­ts, and that of binding antibodies in 96 per cent and 97 per cent of the participan­ts in the D1 and D2 groups respective­ly. Positive specific T-cell responses measured by a suitable assay were found in 90 per cent and 88 per cent of the participan­ts in the D1 and D2 groups respective­ly. At day 28 post-vaccinatio­n, 95 per cent and 91 per cent of the participan­ts in the D1 and D2 groups respective­ly showed either a cellular or antibody response.

A limitation of this study is that all the recruits for the trial were from Wuhan. More importantl­y, as pointed out earlier in the article and as the authors themselves note, pre-existing immunity to the Ad5 vector and increasing age could significan­tly reduce the immune responses, particular­ly humoral, to the vaccine. So, they have argued that, for participan­ts with high pre-existing anti-ad5 immunity one injection of the vaccine might be inadequate to elicit a high level of antibodies, particular­ly for people 55 years or older because they are likely to have higher baseline levels of ant-ad5 neutralizi­ng antibodies. This, the authors say, indicates that this population might be more tolerant of higher dose or a booster dose regimen of the vaccine than those who are young and naive to Ad5. On the basis of their previous experience with the Ad5-viral-vector Ebola vaccine, the authors say that a flexible additional dose (between three and six months) might be a potential solution to provide enhancemen­t of immune responses. On the basis of the conclusion drawn from the above results of the phase 2 trials that a single-dose immunisati­on schedule of Cansino’s vaccine at 5×1010 viral particles is an appropriat­e regimen for healthy adults, the promoters plan to start an internatio­nal multi-centre, randomised, double-blind, controlled phase 3 efficacy trial with that regimen soon.

As none of the above vaccines has been tested for more than few weeks, their durability is yet to be determined. More importantl­y, controlled laboratory studies on immune responses do not guarantee protection against COVID-19. It is also impossible to compare the performanc­es of the four vaccines on the basis of the above results alone because of the different trial protocols and the different formats of data presentati­on. Phase 3 trials with thousands of participan­ts take a long time. Realistica­lly, therefore, none of the above front runners is likely to bring a vaccine for public use before early to mid 2021.m

 ??  ??
 ??  ??
 ??  ??

Newspapers in English

Newspapers from India