MALARIA VACCINES BASED ON PRE-ERYTHROCYTIC ANTIGENS FROM P. FALCIPARUM

The present invention relates to polypeptides or fragments thereof for use as malaria vaccines. It also relates to nucleic acid molecules coding for the polypeptides of the invention. It further relates to compositions comprising such polypeptides or fragments thereof or the nucleic acid molecules, in particular combinations of such polypeptides or fragments thereof, and the use of such compositions as malaria vaccines.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description

The present invention relates to polypeptides or fragments thereof for use as malaria vaccines. It also relates to nucleic acid molecules coding for the polypeptides of the invention. It further relates to compositions comprising such polypeptides or fragments thereof or the nucleic acid molecules, in particular combinations of such polypeptides or fragments thereof, and the use of such compositions as malaria vaccines.

BACKGROUND OF THE INVENTION

Malaria is an infectious disease transmitted by inoculation of infectious sporozoites from the Plasmodium parasite via Anopheles mosquito bites. There are currently an estimated 500 million annual cases of malaria globally, and infection is lethal in 1-3 million people per year in endemic regions such as sub-Saharan Africa. Among the worst affected are children under the age of 5 years, in which the disease comprises several syndromes such as acute respiratory disease, severe anemia or cerebral malaria. The life cycle of Plasmodium falciparum, the parasite causing the most severe form of human malaria, is complex and multifaceted, with local differences within the host tissues and differing morphology corresponding to differential antigen expression. Following sporozoite inoculation, the parasite transforms and expands in an obligate, clinically silent and uni-directional phase in the liver (Prudenco et al., 2006). The release of packages of membrane-enclosed merozoites, termed merosomes, marks the onset of malaria (Sturm et al., 2006) though the symptoms and pathology of malaria, typically recognised by its cyclic pattern of fevers and chills, is caused exclusively during the rapid asexual multiplication phase of the Plasmodium parasite inside erythrocytes (Haldar et al., 2007). Thus, only preventative treatments targeting the pre-erythrocytic stages have the potential to prevent disease and offer effective protection. Immunisation of both humans and rodents with gamma-irradiated P. falciparum sporozoites (RAS) is the only experimental vaccine that confers essentially complete protection and is the gold standard of protective immunity against mosquito-borne malaria transmission (Nussenzweig et al., 1967; Hoffman et al., 2002). Irradiation disrupts the parasite genetic repertoire and parasites arrest in the liver, where they persist for up to 6 months post-infection (Silvie et al., 2002). However, these parasites are genetically undefined and, until today, have been restricted to experimental studies only. Nonetheless, this model provides proof-of-principle that sterile immunity in man is possible, based on the random genetic attenuation obtained by irradiation. Recent work has elicited sterile immunity in rodent models by genetically attenuated parasites (GAPs) (Mueller et al., 2005 a/b; van Dijk et al., 2005; Tarun et al., 2007; Aly et al., 2008; Silvie et al., 2008). These GAPs arrest at the early liver stage and confer sustained and stage-specific immunity (Mueller et al., 2007; Jobe et al., 2007).

The challenge for malaria vaccination based on genetically attenuated parasites depends on translating the results acquired in the rodent malaria models to human malaria. This has been successfully performed by disrupting p52 in P. falciparum, an ortholog of the rodent parasite gene p36p, which has been recently shown to induce sterilising immunity against sporozoite induced malaria (van Schaijk et al., 2008; van Dijk et al., 2005; VanBuskirk et al., 2009). The immune mechanisms conferring resistance to attenuated sporozoites are thought to rely on both humoral and cellular arms of the immune system. Apoptotic RAS-infected hepatocytes lead to presentation of Plasmodium antigens to dendritic cells, likely eliciting the protection-conferring immune response (Leiriao et al., 2005). Primarily, protection has been thought to rely on CD8+ T cells as the cytotoxic effector cells (White et al., 1996; Bongfen et al., 2007) and CD4+ T cells providing aid for antibody and optimal memory CD8+ cytotoxic T lymphocyte activity (Carvalho et al., 2002). Sterile immunity can be achieved in the absence of CD8+ T cells, mediated solely by CD4+ T cells and class-II effector mechanisms (Oliveira et al., 2008). Interestingly, a recently published model has been described where primaquine phosphate, a drug that exclusively targets Plasmodium liver stages, has been utilised in vivo to elicit vaccine-like protective immunity against subsequent sporozoite-induced rodent malaria in the absence of persistent metabolically active liver stages essential for such a CD8+ T cell response (Putrianti et al., 2009). Moreover, the use of wildtype parasites together with prophylactic chemotherapy (such as Chloroquine) results in protective immunity in P. falciparum (Roestenberg et al., 2009). Nonetheless, it has been suggested that long-persisting RAS forms present parasite antigen via MHC class I that induces IFN-gamma-based inducible nitric oxide synthase (iNOS) production by infected hepatocytes (Klotz et al., 1995). The circumsporozoite protein (CSP), the major surface protein of sporozoites, has historically been considered a major antigen and vaccine candidate associated with the liver stages of Plasmodium. Several CSP-derived T cell epitopes have been identified to date, some associated with very high levels of protection, as measured by the reduction in liver stage burden following challenge of immunised mice with normal sporozoites. RTS,S/AS02A is a pre-erythrocytic vaccine candidate based on P. falciparum CSP developed collaboratively between GlaxoSmithKline (GSK) and the Malaria Vaccine Initiative (MVI) Programme for Appropriate Technology in Health (PATH). However, its results are rather disappointing, with efficacies of between 40 and 60% (Alonso et al., 2004; Alonso et al., 2005; Bejon et al., 2008, Epstein et al., 2011). Such partial protection suggests that CSP may not be the sole protective antigen in attenuated models. Indeed, there is no CSP epitope described in C57Bl/6 mice, though in Balb/c there is a well-known and characterised T-cell epitope. Interestingly, a recent study employing a P. berghei parasite line expressing heterologous P. falciparum CSP demonstrated that sterile immunity can be achieved in the absence of a CSP-specific immune response and concluded that hitherto uncharacterised antigens, and not CSP, may be targeted to induce sterilising immunity (Grüner et al., 2007; Mauduit et al., 2010).

It was an object of the present invention to provide means for an effective malaria vaccine. More specifically, it was an object of the present invention to identify novel liver stage antigens that are protective (i.e. critical for immunity), and thus can be used for vaccination.

SUMMARY OF THE INVENTION

According to the present invention this object is solved by a composition comprising at least two polypeptides selected from polypeptides comprising an amino acid sequence of SEQ ID NOs. 1 to 18 and a polypeptide, which is at least 80% identical to any of the above polypeptides,

preferably furthermore selected from polypeptides comprising an amino acid sequence of SEQ ID NOs. 19 to 23 and a polypeptide, which is at least 80% identical to any of the above polypeptides,

or comprising at least two fragments of the above polypeptides.

According to the present invention this object is solved by a composition comprising at least two nucleic acid molecules each coding for at least one polypeptide or fragment thereof according to the invention, preferably having a nucleotide sequence selected from SEQ ID NOs. 41 to 63, more preferably SEQ ID NOs. 41 to 65.

According to the present invention this object is solved by providing the compositions of the invention for use as a malaria vaccine, preferably for use as a subunit malaria vaccine or multicomponent malaria vaccine emulating the malarial live attenuated organisms.

According to the present invention this object is furthermore solved by a polypeptide comprising an amino acid sequence selected from SEQ ID NOs. 1 to 23 and a polypeptide, which is at least 80% identical to any of the above polypeptides, or a fragment thereof, for use as a malaria vaccine, preferably for use as a subunit malaria vaccine or multicomponent malaria vaccine emulating the malarial live attenuated organisms.

According to the present invention this object is furthermore solved by a fragment of the above polypeptide, comprising or having an amino acid sequence selected from SEQ ID NOs. 26 to 36 for use as a malaria vaccine, preferably for use as a subunit malaria vaccine or multicomponent malaria vaccine emulating the malarial live attenuated organisms.

According to the present invention this object is furthermore solved by a nucleic acid molecule coding for at least one polypeptide or fragment thereof according to the invention, preferably having a nucleotide sequence selected from SEQ ID NOs. 41 to 63,

for use as a malaria vaccine, preferably for use as a subunit malaria vaccine or multicomponent malaria vaccine emulating the malarial live attenuated organisms.

DESCRIPTION OF THE PREFERRED EMBODIMENTS OF THE INVENTION

Before the present invention is described in more detail below, it is to be understood that this invention is not limited to the particular methodology, protocols and reagents described herein as these may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention which will be limited only by the appended claims. Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art. For the purpose of the present invention, all references cited herein are incorporated by reference in their entireties.

Concentrations, amounts, and other numerical data may be expressed or presented herein in a range format. It is to be understood that such a range format is used merely for convenience and brevity and thus should be interpreted flexibly to include not only the numerical values explicitly recited as the limits of the range, but also to include all the individual numerical values or sub-ranges encompassed within that range as if each numerical value and sub-range is explicitly recited. As an illustration, a numerical range of “from at least two to 23” should be interpreted to include not only the explicitly recited values of 2 to 23, but also include individual values and sub-ranges within the indicated range. Thus, included in this numerical range are individual values such as 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22 and 23 and sub-ranges such as from 2 to 5, from 3 to 10, from 4 to 15, from 2 to 8, from 5 to 15, and from 6 to 10, etc. As another illustration, a numerical range of “5 to 50 amino acids” should be interpreted to include not only the explicitly recited values of 5 to 50 amino acids, but also include individual values and sub-ranges within the indicated range. Thus, included in this numerical range are individual values such as 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, . . . , 49 and 50 and sub-ranges such as from 5 to 50, from 8 to 25, from 8 to 15, and from 28 to 40, etc. This same principle applies to ranges reciting only one numerical value. Furthermore, such an interpretation should apply regardless of the breadth of the range or the characteristics being described.

Compositions with Malaria Attenuated Liver Stage (MALS) Antigens

Here we present a novel approach for vaccine development that harnesses the immunological potential of the parasite at the liver stage, the clinically silent incubatory state prior to the onset of the pathological blood stage.

The results of the prior art described above in addition to the fact that eventually clinical immunity to malaria is acquired during natural infections suggested that an efficacious vaccine against malaria may in principle be producible. However, while naturally acquired immunity to malaria is believed to target the erythrocytic stage of infection until today only vaccines targeting the pre-erythrocytic stages have resulted in protection against malaria infection. As irradiated sporozoites (attenuated parasites) are difficult to technically produce and given the fact that pre-erythrocytic vaccine candidates based on P. falciparum CSP have been giving disappointing results in terms of efficacy new candidate vaccine antigens are still warranted.

The inventors have found a unique strategy for the identification of such antigens. Now the present invention discloses protective antigens targeting the critical phase of the parasite in the liver. These novel validated antigens isolated from the protection-inducing attenuated parasites—a novelty in malaria vaccine development—can be further translated into a vaccine prototype that mediates immunity by elimination of the parasite at this crucial, pre-pathological bottleneck.

Ultimately, our technology can be further expanded by combining the most potent pre-erythrocytic antigens described within this invention with erythrocytic antigens for a multicomponent (-stage) subunit vaccine that both emulates the live-attenuated parasite vaccine and mimics induction of natural acquired immunity and hence paves the way for the preparation of next generation vaccine formulations.

As described above, the present invention provides a composition comprising:

    • the newly identified MALS antigens (proteins, polypeptides)
    • or
    • fragments thereof.
    • or
    • nucleic acid molecules encoding them.

The compositions according to the invention comprise at least two, at least three, at least four, at least five, at least six, up to 23 (at least 2 to 23) of the polypeptides or nucleic acid molecules encoding them, such as from 2 to 5, from 3 to 10, from 4 to 15, from 2 to 8, from 5 to 15, and from 6 to 10.

The compositions according to the invention comprise fragments of at least two, of at least three, of at least four, of at least five, of at least six, of up to 23 (at least 2 to 23) of the polypeptides of the invention, such as of 2 to 5, of 3 to 10, of 4 to 15, of 2 to 8, of 5 to 15, and of 6 to 10.

The proteins or polypeptides which are to be selected for the composition of the invention are proteins or polypeptides comprising or having an amino acid sequence of

MAL13P1.13 (SEQ ID NO. 1), (MALS_A) PF14_0480 (SEQ ID NO. 2), (MALS_B) MAL13P1.258 (SEQ ID NO. 3), (MALS_C) PF14_0435 (SEQ ID NO. 4), (MALS_F) PF14_0390 (SEQ ID NO. 5), PF13_0242 (SEQ ID NO. 6), PF13_0168 (SEQ ID NO. 7), PF11_0342/0421 (SEQ ID NO. 8), PFc0515c (SEQ ID NO. 9), PFc0135c (SEQ ID NO. 10), PF14_0533 (SEQ ID NO. 11), PF13_0282 (SEQ ID NO. 12), PF10_0081 (SEQ ID NO. 13), PFF0420c (SEQ ID NO. 14), PF14_0323 (SEQ ID NO. 15), (MALS_Cal) PF10_0375 (SEQ ID NO. 16), PF11_0128 (SEQ ID NO. 17), PF14_0548 (SEQ ID NO. 18),

and
    • a polypeptide, which is at least 80%, preferably at least 85%, more preferably at least 90%, even more preferably at least 95% identical to any of the above polypeptides.

They are furthermore selected from proteins or polypeptides comprising or having an amino acid sequence of

PF08_0051 (SEQ ID NO. 19), PF08_0036 (SEQ ID NO. 20), PF13_0135 (SEQ ID NO. 21), PF13_0343 (SEQ ID NO. 22), PFE1260c (SEQ ID NO. 23),

and
    • a polypeptide, which is at least 80%, preferably at least 85%, more preferably at least 90%, even more preferably at least 95% identical to any of the above polypeptides.

For details, see Table 1.

The above accession numbers are PlasmoDB/GeneDB accession numbers (www.plasmodb.org, version: 8.1, October 11, 201; www.genedb.org, version: November, 2010).

As described above, the present invention also provides polypeptides which are at least 80%, preferably at least 85%, more preferably at least 90%, even more preferably at least 95% identical to any of the above polypeptides, or fragments thereof.

As used herein, the term “percent (%) identical” or “percent (%) sequence identity” refers to sequence identity between two amino acid sequences. Identity can be determined by comparing a position in both sequences, which may be aligned for the purpose of comparison. When an equivalent position in the compared sequences is occupied by the same amino acid, the molecules are considered to be identical at that position.

Generally, a person skilled in the art is aware of the fact that some amino acid exchanges in the amino acid sequence of a protein or peptide do not have any influence on the (secondary or tertiary) structure, function and activity of the protein or peptide at all. Amino acid sequences with such “neutral” amino acid exchanges as compared to the amino acid sequences disclosed herein fall within the scope of the present invention. Also included are mutations in the original amino acid sequences that allow or facilitate the production of the polypeptide or fragments thereof in an organism different from P. falciparum, such as E. coli.

Preferably, the composition of the invention comprises at least two polypeptides which are selected from polypeptides comprising or having an amino acid sequence of SEQ ID NOs. 1 to 23 or polypeptides having at least 80% sequence identity to any of the amino acid sequences of SEQ ID NOs. 1 to 23 or fragment(s) thereof.

In a preferred embodiment, a composition of the invention furthermore comprises

    • P. falciparum Ferlin,
    • P. falciparum Ferlin-like protein and/or
    • another P. falciparum C2-domain containing protein, or
    • or a fragment thereof.

P. falciparum Ferlin (PfFER; PF140530) has the amino acid sequence of SEQ ID NO. 24, P. falciparum Ferlin-like protein (Pf FLP; MAL8P 1.134) has the amino acid sequence of SEQ ID NO. 25.

A composition of the invention can furthermore comprise polypeptide(s) comprising or having an amino acid sequence selected from

PF14_0530 (SEQ ID NO. 24), (MALS_E), Mal8P1.134 (SEQ ID NO. 25), (MALS_G),

and

polypeptides having at least 80% sequence identity to any of the amino acid sequences of SEQ ID NOs. 24 and 25, or fragment(s) thereof.

Preferably, the composition of the invention comprises at least two polypeptides which are selected from polypeptides comprising or having an amino acid sequence of SEQ ID NOs. 1 to 25 or polypeptides having at least 80% sequence identity to any of the amino acid sequences of SEQ ID NOs. 1 to 25 or fragment(s) thereof.

Preferred Compositions

A preferred composition of the invention comprises at least two of

MAL13P1.13 (SEQ ID NO. 1), (MALS_A), PF14_0480 (SEQ ID NO. 2), (MALS_B), MAL13P1.258 (SEQ ID NO. 3), (MALS_C), PF14_0435 (SEQ ID NO. 4), (MALS_F),

and polypeptides having at least 80% sequence identity to any of the amino acid sequences of SEQ ID NOs. 1 to 4, or fragment(s) thereof.

A preferred composition of the invention comprises at least two of

MAL13P1.13 (SEQ ID NO. 1), (MALS_A), PF14_0480 (SEQ ID NO. 2), (MALS_B), MAL13P1.258 (SEQ ID NO. 3), (MALS_C), PF14_0435 (SEQ ID NO. 4), (MALS_F), PF14_0530 (SEQ ID NO. 24), (MALS_E), PF14_0323 (SEQ ID NO. 15), (MALS_Cal)

and polypeptides having at least 80% sequence identity to any of the amino acid sequences of SEQ ID NOs. 1 to 4, 15 or 24, or fragment(s) thereof.

Fragments

In an embodiment of the invention, the composition comprises fragments of the polypeptides of the invention.

Preferably, a fragment of the polypeptide(s) of the invention, namely the polypeptides comprising or having the amino acid sequence of SEQ ID NOs. 1 to 23 (or SEQ ID NOs. 1 to 25), comprises at least one antigenic determinant or epitope of the polypeptide.

In one embodiment, the amino acid sequence of the antigenic determinant or epitope has a length of at least 8 amino acids. Thus, in a preferred embodiment, the fragment has a length of at least 8 amino acids. In an embodiment, the fragment or peptide has a length in the range of 5 to 50 amino acids, preferably 8 to 25 amino acids, more preferably 8 to 15 amino acids. In an embodiment, the fragment or peptide has a length in the range of 28 to 40 amino acids.

In one embodiment, the antigenic determinant or epitope is a CD8+ T cell epitope, a CD4+ T cell epitope or a B cell epitope, preferably a CD8+ T cell epitope. Preferably, the CD8+ T cell epitope is a P. falciparum-specific CD8+ T cell epitope, such as a HLA-A 0201-restricted CD8+ T cell epitope. A person skilled in the art knows how to identify/predict CD8+ T cell epitopes in a given amino acid sequence, e.g. by epitope prediction programs, such as SYFPEITHI (http://www.syfpeithi.de).

In a preferred composition (of at least two fragments) the fragments are selected from fragments comprising or having an amino acid sequence of

(SEQ ID NO. 26) SLICGLYLL, (fragment of MALS_A), (SEQ ID NO. 27) ILYSLMINSL, (fragment of MALS_A), (SEQ ID NO. 28) LICGLYLLTL, (fragment of MALS_A), (SEQ ID NO. 29) VLLEKINVI, (fragment of MALS_B), (SEQ ID NO. 30) YLSPNFINKI, (fragment of MALS_B), (SEQ ID NO. 31) ILHGGVYRL, (fragment of MALS_C), (SEQ ID NO. 32) ILFLFILSI, (fragment of MALS_C), (SEQ ID NO. 33) LLFINEINKL, (fragment of MALS_C), (SEQ ID NO. 34) SLISLYIYYV, (fragment of MALS_F), (SEQ ID NO. 35) FLLLMLVSI, (fragment of MALS_F), and/or (SEQ ID NO. 36) FLTLMARKL. (fragment of MALS_Cal).

They can furthermore be selected from fragments comprising or having an amino acid sequence of

(SEQ ID NO. 37) NLLDPLVVV, (fragment of MALS_E), (SEQ ID NO. 38) LLLEGNFYL, (fragment of MALS_E), (SEQ ID NO. 39) KLIPVNYEL, and (fragment of MALS_E), and/or (SEQ ID NO. 40) ILIPSLPLI. (fragment of MALS_E).

In a preferred composition (of at least two fragments) the fragments are selected from fragments of MALS_A (SEQ ID NO. 1), MALS_B (SEQ ID NO. 2), MALS_C (SEQ ID NO. 3), MALS_F (SEQ ID NO. 4) and optionally MALS_Cal (SEQ ID NO. 15) and MALS_E (SEQ ID NO. 24).

The following mixtures/pools of fragments (peptide pools of antigens) are preferred:

    • Fragments of MALS_A SEQ ID NOs. 26 to 28,
    • Fragments of MALS_B SEQ ID NOs. 29 and 30,
    • Fragments of MALS_C SEQ ID NOs. 31 to 33,
    • Fragments of MALS_F SEQ ID NOs. 34 and 35,
    • Fragments of MALS_Cal SEQ ID NO. 36,
    • Fragments of MALS_E SEQ ID NOs. 37 to 40.

A preferred composition comprises fragments comprising or having an amino acid sequence of

    • SEQ ID NOs. 26 to 28, and/or
    • SEQ ID NOs. 29 and 30, and/or
    • SEQ ID NOs. 31 to 33, and/or
    • SEQ ID NOs. 34 and 35, and/or
      preferably furthermore
    • SEQ ID NO. 36, and/or
    • SEQ ID NOs. 37 to 40.

The use of antigenic peptides of Ferlin, Ferlin-like protein and other C2-domain containing proteins for malaria vaccines is also disclosed in WO 2011/066995.

Modifications

In one embodiment, the polypeptide(s) of the invention or fragment(s) thereof comprise further component(s), such as label(s), N- and/or C-terminal modification(s), further drug(s) or agent(s). The skilled artisan will be able to select suitable further components.

Nucleic Acid Molecules Coding for the Polypeptide(s) of the Invention

In an embodiment of the invention, the composition comprises nucleic acid molecules coding for the polypeptides of the invention.

Preferred nucleic acid molecules have a nucleotide sequence selected from SEQ ID NOs. 41 to 63 or SEQ ID NOs. 41 to 65.

A preferred composition of the invention comprises at least two nucleic acid molecules each coding for at least one polypeptide of the invention or fragment thereof, preferably having a nucleotide sequence selected from SEQ ID NOs. 41 to 63, more preferably SEQ ID NOs. 41 to 65.

The nucleic acid molecules can be isolated nucleic acid molecules, plasmids comprising them, expression constructs comprising them, such as expression systems for DNA vaccines.

Further Components of the Compositions

The compositions according to the invention can further comprise

    • a carrier
    • and/or
    • an adjuvant.

In one embodiment, the carrier is fused to the polypeptide(s) or fragment(s) thereof or nucleic acid molecule(s) encoding them (i.e. the components of the composition as described above).

In one embodiment, the carrier is a virus particle or parts thereof, an envelope protein of a viral vector or of a virus particle, a nanocarrier.

In one embodiment, the virus particle is Hepatitis B virus particle or parts thereof.

In such an embodiment, the carrier, e.g. Hepatitis B virus particle or parts thereof, is suitable for liver targeting of the polypeptide or fragment thereof.

In one embodiment, the nanocarrier is a cell-targeted nanocarrier, such as the cell-targeted nanocarriers available from Rodos BioTarget GmbH, Hannover (www.biotargeting.org), e.g. the TargoSphere® delivery system. These nanocarriers can be combined with the desired polypeptide or fragment thereof and specifically directed to antigen-presenting immuno cells (like APCs, DCs, macrophages etc).

In one embodiment, the adjuvant is triggering CD8 T cell responses in general. Preferably, the adjuvant is a commercially available adjuvant system, e.g. IC31 (Intercell company, Vienna) since that adjuvant system is triggering CD8 T cell responses rather than antibody-mediated immunity.

Use of the Compositions as Malaria Vaccine

As described above, the present invention provides the compositions for use as a malaria vaccine.

The compositions are preferably for use as a subunit malaria vaccine or multicomponent malaria vaccine emulating the malarial live attenuated organisms.

In comparison to a vaccine based on attenuated whole organisms (live attenuated vaccines), a “subunit vaccine” consists of at least one component (protein, protein fragment) of the respective pathogen.

A “multicomponent vaccine” refers to a vaccine comprising more than one component (protein, protein fragment) of the respective pathogen, preferably from different developmental and disease-inducing life cycle stages (multi-component (-stage) vaccine).

A “subunit vaccine emulating the malarial live attenuated organisms” refers to the minimal combination of immunogenic protein components derived from the protection-inducing attenuated malarial liver stage parasite.

As discussed above, the invention presents a novel approach for vaccine development that harnesses the immunological potential of the parasite at the liver stage, the clinically silent incubatory state prior to the onset of the pathological blood stage.

The herein disclosed and newly identified protective antigens target the critical phase of the parasite in the liver. These novel validated antigens isolated from the protection-inducing attenuated parasites—a novelty in malaria vaccine development—can be further translated into a vaccine prototype that mediates immunity by elimination of the parasite at this crucial, pre-pathological bottleneck.

The present invention can be further expanded by combining the most potent pre-erythrocytic antigens described within this invention with erythrocytic antigens for a multi-component (-stage) subunit vaccine that both emulates the live-attenuated parasite vaccine and mimics induction of natural acquired immunity and hence paves the way for the preparation of next generation vaccine formulations.

MALS Antigens, Fragments, Nucleic Acid Molecules as Targets for Malaria Vaccines

As described above, the present invention furthermore provides a polypeptide comprising or having an amino acid sequence selected from

MAL13P1.13 (SEQ ID NO. 1), (MALS_A) PF14_0480 (SEQ ID NO. 2), (MALS_B) MAL13P1.258 (SEQ ID NO. 3), (MALS_C) PF14_0435 (SEQ ID NO. 4), (MALS_F) PF14_0390 (SEQ ID NO. 5), PF13_0242 (SEQ ID NO. 6), PF13_0168 (SEQ ID NO. 7), PF11_0342/0421 (SEQ ID NO. 8), PFc0515c (SEQ ID NO. 9), PFc0135c (SEQ ID NO. 10), PF14_0533 (SEQ ID NO. 11), PF13_0282 (SEQ ID NO. 12), PF10_0081 (SEQ ID NO. 13), PFF0420c (SEQ ID NO. 14), PF14_0323 (SEQ ID NO. 15), (MALS_Cal) PF10_0375 (SEQ ID NO. 16), PF11_0128 (SEQ ID NO. 17), PF14_0548 (SEQ ID NO. 18), PF08_0051 (SEQ ID NO. 19), PF08_0036 (SEQ ID NO. 20), PF13_0135 (SEQ ID NO. 21), PF13_0343 (SEQ ID NO. 22), PFE1260c (SEQ ID NO. 23),

and
    • a polypeptide, which is at least 80%, preferably at least 85%, more preferably at least 90%, even more preferably at least 95% identical to any of the above polypeptides,
      or a fragment thereof,

for use as a malaria vaccine, preferably for use as a subunit malaria vaccine or multicomponent malaria vaccine emulating the malarial live attenuated organisms.

As described above, the present invention furthermore provides a fragment of a polypeptide of the invention, comprising or having an amino acid sequence selected from

(SEQ ID NO. 26) SLICGLYLL, (fragment of MALS_A), (SEQ ID NO. 27) ILYSLMINSL, (fragment of MALS_A), (SEQ ID NO. 28) LICGLYLLTL, (fragment of MALS_A), (SEQ ID NO. 29) VLLEKINVI, (fragment of MALS_B), (SEQ ID NO. 30) YLSPNFINKI, (fragment of MALS_B), (SEQ ID NO. 31) ILHGGVYRL, (fragment of MALS_C), (SEQ ID NO. 32) ILFLFILSI, (fragment of MALS_C), (SEQ ID NO. 33) LLFINEINKL, (fragment of MALS_C), (SEQ ID NO. 34) SLISLYIYYV, (fragment of MALS_F), (SEQ ID NO. 35) FLLLMLVSI, (fragment of MALS_F), and/or (SEQ ID NO. 36) FLTLMARKL. (fragment of MALS_Cal),

for use as a malaria vaccine, preferably for use as a subunit malaria vaccine or multicomponent malaria vaccine emulating the malarial live attenuated organisms

As described above, the present invention furthermore provides a nucleic acid molecule coding for at least one polypeptide of the invention or fragment thereof, preferably having a nucleotide sequence selected from SEQ ID NOs. 41 to 63, for use as a malaria vaccine, preferably for use as a subunit malaria vaccine or multicomponent malaria vaccine emulating the malarial live attenuated organisms.

In another aspect, the present invention relates to a plasmid comprising at least one nucleic acid molecule or sequence as defined above.

In another aspect, the present invention relates to an antibody against a polypeptide or fragment thereof as defined above.

In another aspect, the present invention relates to a method of producing a composition as defined above comprising the step of admixing

at least two polypeptides or fragments thereof as defined above or at least two nucleic acid molecules as defined above.

In yet another aspect, the present invention relates to a method of prevention of malaria, comprising the step of administering a polypeptide or fragment thereof as defined above, or a nucleic acid molecule or plasmid as defined above, or a composition as defined above to a person in need thereof.

TABLE 1 Overview of the 24 most abundantly upregulated transcripts in malaria attenuated liver stages (MALS). SEQ p- ID Gene P. falciparum P. berghei P. yoelii Function value NO. 1 PF13_0242 PBANKA_135860 PY02505 isocitrate 1.6e−32 6 dehydrogenase, mitochondrium 2 PF14_0480 PBANKA_131420 PY01987 C-terminal TM, 0.0001 2 ARM 3 PF14_0530 PBANKA_131930 PY05745 Ferlin, putative 0.0001 24 4 PF13_0168 PBANKA_134630 PY04297 CPW-WPC family 0.0001 7 protein, apicoplast 5 Mal13P1.13 PBANKA_140100 PY02854 Cytoplasmic, 0.0001 1 transcription, SEN-1 related 6 PF11_0342/0421 PBANKA_091520 PY01035 Actin binder 0.0007 8 (PfEMP3) 7 PFc0515c/0135c PBANKA_041020 PY01708 TPR domain 0.001 9, 10 (ARM) containing protein 8 Mal13P1.258 PBANKA_136400 none Extracellular, 0.0011 3 metabolic carbohydrates 9 PF14_0533 PBANKA_131970 PY00668 ApiAP2, apicoplast 0.0011 11 10 PF14_0435 PBANKA_130960 PY05937 Membrane protein, 0.0012 4 apicoplast 11 PF13_0282 PBANKA_113040 PY02352 Proteasome 0.0012 12 12 PF10_0081 PBANKA_120660 PY00768 26S Proteasome 0.0013 13 subunit 13 PFF0420c PBANKA_010710 PY03034 20S Proteasome 0.0013 14 14 Mal8P1.134 PBANKA_122440 PY04695 Ferlin like protein, 0.0013 25 putative 15 PF14_0323 PBANKA_101060 PY06908 Calmodulin 0.0013 15 16 PF10_0375 none none Plasmodium exported 0.0023 16 protein 17 PF11 _0128 PBANKA_093540 PY05764 Coq4 homolog, 0.005 17 Proteasome 18 PF08_0051 PBANKA_071150 none TNF-like domain 0.0057 19 19 PF08_0036 PBANKA_070800 PY02497 Pfsec23 0.007 20 20 PF14_0548 PBANKA_132120 PY00672 PfVps4, AAA 0.0076 18 domain 21 PF13 _0135 PBANKA_133900 PY06640 vacuolar protein 0.008 21 sorting 52 homologue, 2TM domains 22 PF13_0343 PBANKA_114110 none cytosolic 0.012 22 23 PFE1260c PBANKA_124010 PY06789 MAL5P1.252, TM 0.041 23 24 PF14 _0390 none none Transcription, 0.07 5 membrane

The following examples and drawings illustrate the present invention without, however, limiting the same thereto.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 shows the experimental setup of the modified suppression subtractive hybridisation (SSH) assay for comparing the transcripts of P. falciparum WT and RAS parasites after mid-liver stage development.

FIG. 2 shows the predicted primary structure of the indicated P. falciparum proteins, as predicted by the SMART algorithm (www.smart.embl-heidelberg.de, version 7.0, November 2011), Pfam, EMBL and PlasmoDB.org.

FIG. 3. Validation of upregulation of RAS-specific antigens by quantitative RT-PCR. Total RNA has been isolated from P. falciparum liver stages (2 days post infection) of Wildtype and RAS followed by first strand cDNA synthesis. cDNA was then subjected for gene-specific qRT-PCR. Statistical analysis was performed by t-test.

FIG. 4. Cultured ELISpot analysis for detecting antigen-specific CD8+ T cells in malaria-exposed Kenyan adults.

Freshly isolated PBMCs from the blood of malaria-exposed (13 blood samples) and non-exposed (naïve) individuals (8 blood samples) were stimulated with peptide pools of antigen

(A) MALS_A (Mal13P1.13) (SLICGLYLL, ILYSLMINSL, LICGLYLLTL),

(B) MALS_B (PF140480) (VLLEKINVI, YLSPNFINKI),

(C) MALS_C (Mal13P1.258) (ILHGGVYRL, ILFLFILSI, LLFINEINKL),

(D) MALS_F (PF140435) (FLLLMLVSI, SLISLYIYYV).

For statistical analysis we performed a Mann-Whitney U-Test.

FIG. 5. Cultured ELISpot analysis for detecting antigen-specific CD8+ T cells in malaria-exposed Ghanian adults.

Freshly isolated PBMCs from the blood of malaria-exposed (26 blood samples) and non-exposed (naïve) individuals (8 blood samples) were stimulated with peptide pools of antigen

(A) MALS_A (Mal13P1.13) (SLICGLYLL, ILYSLMINSL, LICGLYLLTL),

(B) MALS_B (PF140480) (VLLEKINVI, YLSPNFINKI),

(C) MALS_Cal (PF140323) (FLTLMARKL) as well as peptides derived from the described blood-stage antigen MSP1 (D) (merozoite surface protein 1) (YLIDGYEEI, KLLDKINEI, KLKEFIPKV).

The secretion of IFN-gamma has been analysed by cultured ELISpot assay. For statistical analysis we performed a Mann-Whitney U-Test.

FIG. 6. Cultured ELISpot analysis for detecting antigen-specific CD8+ T cells in malaria-exposed Ghanian adults.

Freshly isolated PBMCs from the blood of malaria-exposed (26 blood samples) and non-exposed (naïve) individuals were stimulated with peptide pools of antigen

(A) MALS_C (Mal13P1.258) (ILHGGVYRL, ILFLFILSI, LLFINEINKL),

(B) MALS_F (PF140435) (SLISLYIYYV),

(C) MALS_E (NLLDPLVVV, LLLEGNFYL, KLIPVNYEL, ILIPSLPLI),

(D) Antigen mixture (MALS_A: SLICGLYLL, ILYSLMINSL, LICGLYLLTL; MALS_B: VLLEKINVI, YLSPNFINKI; MALS_C: ILHGGVYRL, ILFLFILSI, LLFINEINKL; MALS_E: NLLDPLVVV, LLLEGNFYL, KLIPVNYEL, ILIPSLPLI; MALS_F: SLISLYIYYV; MALS_Cal: FLTLMARKL) as well as peptides derived from

(E) MSP1 (merozoite surface protein 1) a described blood-stage antigen (YLIDGYEEI, KLLDKINEI, KLKEFIPKV).

The secretion of IFN-gamma has been analysed by a cultured ELISpot assay. For statistical analysis we performed a Mann-Whitney U-Test.

EXAMPLES Example 1 Identification of Potentially Immunogenic Antigens in the Liver Stages of P. falciparum by a SSH Screen

It has been shown that immunisation with attenuated parasites confer sterile protection in mice (RAS and GAP) and also in humans (RAS). By applying the SSH (suppressive subtractive hybridisation) technology in order to analyse differentially expressed genes the inventors identified critical/potential targets of protective liver-stage immunity. For that purpose, they compared the transcriptional profile of liver stages from wildtype and attenuated (RAS) parasites, i.e. the cDNA populations of the protected RAS forms and the unprotected WT forms. Their modified suppressive subtractive hybridisation (SSH) screening (FIG. 1) allowed selective enrichment of differentially regulated cDNAs of high and low abundance that are exclusively present in one population. A combination of hybridisation and PCR amplification steps allowed simultaneous normalisation and subtraction of the cDNA populations. Since protective antigens are expressed very early, malaria infected hepatocytes were harvested at early time points after host cell invasion. SMART PCR cDNA Synthesis and PCR-Select cDNA subtraction kits were used to identify differentially expressed genes between the two populations.

Sequencing of the identified differentially expressed genes was conducted by a sequencing company GATC, Konstanz and the data obtained were evaluated with BLAST algorithms (PlasmoDB version: 8.1, Oct. 8, 2011, NCBI, Sanger/GeneDB as of November, 2010, SMART). After sequencing and bioinformatical analysis of 672 RAS-specific clones the inventors were able to describe the 24 most abundantly transcribed antigens as listed in Table 1.

Their predicted primary structures are shown in FIG. 2.

Example 2 Validation of Upregulation of RAS-Specific Antigens by Quantitative RT-PCR

The results obtained from the differential expression analyses (Suppression Subtractive Hybridisation screening) have been validated by performing quantitative Real-time PCR (qRT-PCR) for selected antigens. Total RNA isolated from P. falciparum wildtype and radiation-attenuated (RAS) liver stage parasites have been used to quantify the respective transcripts in both populations. Plasmodium liver stages were obtained by infecting cultured primary human hepatocytes with P. falciparum salivary gland sporozoites (strain NF54, Prof Robert Sauerwein, Nijmegen, Netherlands; Delemarre B J M & Van der Kaay H J, Ned. T. Geneesk 123 (1979).

Infection of host cells was conducted in close collaboration with the laboratory of Prof D. Mazier at INSERM, Paris (Semblat et al., 2002). Purified human hepatocytes were plated on collagen I-coated wells (2.5×106 cells/well in a 6-well plate) and maintained until infection with salivary gland sporozoites. Cells have been infected with 1.5×106 sporozoites per well. To obtain liver stages from radiation-attenuated parasites, sporozoites were irradiated at 150 Gray prior infection of liver cells. Two days post infection cells were harvested and total RNA has been purified using TRIZOL. First strand cDNA synthesis (Superscript III 1st strand synthesis Kit, Invitrogen) was performed with gene-specific primers (Invitrogen) in a nested first-strand reaction.

Subsequently, cDNA was used to amplify the corresponding transcripts. SYBR Green has been used for quantification (Power SYBR Green Mastermix, Applied Biosystems). For normalisation, parasite specific GAPDH has been used. We also included as a positive control in this analysis the candidate antigens Pf Ferlin (PF140530; MALS_E) and Pf Ferlin-like protein (Mal8P1.134; MALS_G) (as described and disclosed in WO 2011/066995).

The following oligonucleotides have been used for the nested first strand cDNA synthesis:

Pf GAPDH nested rev [SEQ ID NO. 66] 5′ CAGTGGATGCATGAACGGTGG, MALS_B( PF14_840) nested rev [SEQ ID NO. 67] 5′ CCTAACTTGGAACATGGGAGTC, MALS_A (Mal13P1.13) nested rev [SEQ ID NO. 68] 5′ CTGCACTCTTCCAAAGCCATG, MALS_C (Mal13P1.258) nested rev [SEQ ID NO. 69] 5′ ACCATCGTCTTTACCGTGTGAC, MALS_F (PF14_0435) nested rev [SEQ ID NO. 70] 5′ CTCACGACATTCGAAATGTAATCTC, MALS_E (PF14_0530) nested rev [SEQ ID NO. 71] 5′ GATCATCATGTTGTTTGAATGATTATACC, MALS_G (Mal8P1.134) nested rev [SEQ ID NO. 72] 5′ CATAATCGAAGCCGTTGCAGC.

The following oligonucleotides have been used for amplification of the corresponding transcripts:

Pf GAPDH for [SEQ ID NO. 73] 5′ GCAGCCTTTGGAAGGAAAGAand Pf GAPDH rev [SEQ ID NO. 74] 5′ GGCTCCTCCCTTAAGGTGAC, MALS_B (PF14_0840) for [SEQ ID NO. 75] 5′ CGTGCAGCTCTTTAGTAGAAGTGG and MALS_B rev [SEQ ID NO. 76] 5′ AGCATTAACAGCAGGGTAACTG, MALS_A (Mal13P1.13) for [SEQ ID NO. 77] 5′ TATCGTGCACATATGACCATCT and MALS_A rev [SEQ ID NO. 78] 5′ CATCTCCCTTGTCCATTTGCAAC, MALS_C (Mal13P1.258) for [SEQ ID NO. 79] 5′ GGTCTCAGGTATGGACAGGG and MALS_C rev [SEQ ID NO. 80] 5′ TCATGATCAGGATGGGGAGATG, MALS_F ( PF14_0435) for [SEQ ID NO. 81] 5′ CGACAAATACATAAAGATGGACGAG and MALS_F rev [SEQ ID NO. 82] 5′ CATGGCTTGTTGGTATAAAACATACG, MALS_E (PF14_0530) for [SEQ ID NO. 83] 5′ GCAGCTCTCGTCATATCAGCA and MALS_E rev [SEQ ID NO. 84] 5′ TCCAAGCTTCGTCATCATCGT, MALS_G (Mal8P1.134) for [SEQ ID NO. 85] 5′ GAGCCTATAGGTGAGGCAACC and MALS_G rev [SEQ ID NO. 86] 5′ CCAACTGGGTCAAGTTCAGCC.

The quantification of the transcript copies clearly shows a 1.2 to 6.5-fold up-regulation of the tested antigens in liver stages of radiation-attenuated parasites compared to wildtype liver stages (see FIG. 3).

Example 3 Presence of Antigen-Specific T Cells Recognising Peptides Derived from Selected Antigen Candidates in Malaria-Exposed Kenyan Adults

In order to investigate the presence of antigen-specific T cells in malaria-exposed individuals, T-cell responses to peptides from a first selection of antigens MAL13P1.13 (SLICGLYLL, ILYSLMINSL, LICGLYLLTL), PF140480 (VLLEKINVI, YLSPNFINKI), MAL13P1.258 (ILHGGVYRL, ILFLFILSI, LLFINEINKL), and PF140435 (FLLLMLVSI, SLISLYIYYV) were tested in semi-immune Kenyan adults in collaboration with Dr. Britta Urban at the Kenyan Medical Research Institute-Wellcome Trust Research Programme (KEMRI). All adults are resident in Junju District, about 60 km north of Mombasa at the Kenyan coast. The area has two high transmission seasons but low-level transmission occurs all year round (infectious bites per year: 23-53) (Mwangi et al., 2005).

In order to determine the production of antigen-specific IFN-gamma by activated peripheral blood mononuclear cells (PBMC), cultured ELISpot analysis was carried out over a period of 10 days. Peripheral blood mononuclear cells (PBMCs) were purified from fresh blood samples by gradient centrifugation using Lymphoprep and resuspended in RPMI 1640 medium containing 10% heat-inactivated FCS, 2 mM L-Glutamine and Penicillin (100 U/ml)/Streptomycin (100 μg/ml). 1×106 cells were cultured with 10 μg/ml of peptides in a volume of 500 μl. On day 3 and day 7, 250 μl culture medium was removed and replaced with fresh medium containing human IL2 (final concentration 20 U/ml). On day 9, cells were washed three times, resuspended in 500 μl medium and rested overnight before proceeding to an IFN-gamma ELISpot assay (IFN-gamma ELISpot kit, Mabtech). Cells (100.000 cells/well) were transferred to MultiScreen filter plates coated with 10 μg/ml anti-human IFN-gamma antibody and incubated with indicated peptide pools or medium (non stimulated control) overnight (in each case in triplicates). After removing the cells and several washing steps, secreted IFN-gamma was detected by using a second antibody against human IFN-gamma coupled with biotin (1 μg/ml) and subsequent addition of streptavidin-ALP followed by substrate solution.

The detected IFN-gamma response is shown as counted spots per million cells. FIG. 4 summarises the specific T-cell responses to individual peptide pools from antigens Mal13P1.13, PF140840, Mal13P1.258 (high responder) as well as PF140435 (mediate responder), which therefore can be considered as valuable candidate antigens for a potential subunit vaccine against malaria.

Example 4 Presence of Antigen-Specific T Cells Recognizing Peptides Derived from Selected Antigen Candidates in Semi-Immune Ghanaian Adults

We have been able to expand our investigations on testing reactivity of T cells to our critical target antigens in a malaria holo-endemic region in Ghana.

In close collaboration with Prof. Dr. Achim Hoerauf (University of Bonn, Germany) and the Kumasi Center for Collaborative Research, Kumasi, Ghana (KCCR), we investigated the presence of antigen-specific T cells recognising peptides derived from selected antigen candidates in semi-immune Ghanaian adults. Most of the analysed peptides have already been tested in studies in Kilifi, Kenya as described as part of this invention. Within this study the following antigen candidates and corresponding peptides were used: MALS_A=Mal13P1.13 (SLICGLYLL, ILYSLMINSL, LICGLYLLTL), MALS_B=PF140840 (VLLEKINVI, YLSPNFINKI), MALS_C=Mal13P1.258 (ILHGGVYRL, ILFLFILSI, LLFINEINKL), MALS_F=PF140435 (SLISLYIYYV) and MALS_Cal ═PF140323 (FLTLMARKL). In addition to that we included peptides derived from the candidate antigen Pf Ferlin (PF140530) as described below (and disclosed in WO 2011/066995).

In order to determine the production of antigen-specific IFN-gamma by activated peripheral blood mononuclear cells (PBMC), cultured ELISpot analyses were carried out over a period of 10 days. Briefly, Peripheral Blood Mononuclear Cells (PBMCs) were purified from fresh blood samples by gradient centrifugation using Lymphoprep and resuspended in RPMI 1640 medium containing 10% heat-inactivated FBS, 2 mM L-Glutamine and Penicillin (100 U/ml)/Streptomycin (100 μg/ml). 1×106 cells were cultured with 10 μg/ml (except for the tested antigen mixture, described below) of peptides in a volume of 8000. On day 3 and day 7, 400 μl culture medium was removed and replaced with fresh medium containing human IL2 (final concentration 20 U/ml). On day 9 cells were washed three times, resuspended in 500 μl medium and rested overnight before proceeding with an IFN-gamma ELISpot assay (IFN-gamma ELISpot kit, Mabtech). Cells (100.000 cells/well) were transferred to MultiScreen filter plates coated with 10 μg/ml anti-human IFN-gamma antibody and incubated with indicated peptide pools or medium (non stimulated control) overnight (in each case in triplicates). After removing the cells and several washing steps, secreted IFN-gamma was detected by using a secondary antibody against human IFN-gamma coupled with biotin (1 μg/ml) and subsequent addition of Streptavidin-ALP followed by substrate solution. The detected IFN-gamma response is shown as counted spots per million cells. In addition to analysing peptide pools from individual antigens, we investigated T cell activation in response to a mixture of antigens (peptide pool). In this experimental set-up, peptides derived from antigen candidates Mal13P1.13 (SLICGLYLL, ILYSLMINSL, LICGLYLLTL), PF140840 (VLLEKINVI, YLSPNFINKI), Mal 13P1.258 (ILHGGVYRL, ILFLFILSI, LLFINEINKL), PF140435 (SLISLYIYYV), PF140323 (FLTLMARKL) and PF140530 (NLLDPLVVV, LLLEGNFYL, KLIPVNYEL, ILIPSLPLI) were combined at a concentration of 1.25 μg/ml per peptide (considered as peptide pool).

Two different sets of experiments have been carried out using 26 blood samples of malaria-exposed individuals (semi-immune adults) each (FIG. 5 and FIG. 6, respectively). In parallel CD8+ T cell responses to peptides derived from a known blood-stage vaccine candidate MSP-1 (Merozoite Surface Protein 1) have been determined (Goodman et al., 2010). In good agreement with our hypothesis that parasite transcripts isolated from the protection-mediated attenuated parasite line may serve as critical targets of anti-liver stage immunity we could nicely confirm a significant interaction of these candidate antigens with the host's immune system. Cultured ELISpot analysis hence revealed significant T cell activation as measured by IFN-gamma secretion for the target antigens MALS_A (Mal13P1.13), MALS_C (Mal13P1.258), MALS_E (PF140530) and MALS_F (PF140435).

With respect to the results shown in FIG. 5: Contrary to what was expected we measured IFN-gamma responses against MALS_A in a few individuals from the non-exposed group. We excluded specific restimulation of PBMCs from non-exposed individuals by MALS_A derived peptides after BLAST searches of the specific peptide sequence against human and other apicomplexan parasites (including Toxoplasma) sequences. Evidence from the literature suggests that Plasmodium antigens are thought to mimic antigens from other microbes thereby soliciting the reactivation of cross-reactively primed memory T cells rapid (Riley et al, 1999).

The features disclosed in the foregoing description, in the claims and/or in the accompanying drawings may, both separately and in any combination thereof, be material for realizing the invention in diverse forms thereof.

REFERENCES

  • Alonso, P. L., Sacarlal, J., Aponte, J. J., Leach, A., Macete, E., Milman, J., Mandomando, I., Spiessens, B., Guinovart, C., Espasa, M., Bassat, Q., Aide, P., Ofori-Anyinam, O., Navia, M. M., Corachan, S., Ceuppens, M., Dubois, M. C., Demoitie, M. A., Dubovsky, F., Menendez, C., Tornieporth, N., Ballou, W. R., Thompson, R., Cohen, J. (2004) Efficacy of the RTS,S/AS02A vaccine against Plasmodium falciparum infection and disease in young African children: randomised controlled trial. Lancet 364, 1411-20.
  • Alonso, P. L., Sacarlal, J., Aponte, J. J., Leach, A., Macete, E., Aide, P., Sigauque, B., Milman, J., Mandomando, I., Bassat, Q., Guinovart, C., Espasa, M., Corachan, S., Lievens, M., Navia, M. M., Dubois, M. C., Menendez, C., Dubovsky, F., Cohen, J., Thompson, R., Ballou, W. R. (2005) Duration of protection with RTS,S/AS02A malaria vaccine in prevention of Plasmodium falciparum disease in Mozambican children: single-blind extended follow-up of a randomised controlled trial. Lancet 366, 2012-8.
  • Aly, A. S., Mikolajczak, S. A., Rivera, H. S., Camargo, N., Jacobs-Lorena, V., Labaied, M., Coppens, I., Kappe, S. H. (2008) Targeted deletion of SAP1 abolishes the expression of infectivity factors necessary for successful malaria parasite liver infection. Mol Microbiol. 69, 152-63.
  • Bejon, P., Lusingu, J., Olotu, A., Leach, A., Lievens, M., Vekemans, J., Mshamu, S., Lang, T., Gould, J., Dubois, M. C., Demoitie, M. A., Stallaert, J. F., Vansadia, P., Carter, T., Njuguna, P., Awuondo, K. O., Malabeja, A., Abdul, O., Gesase, S., Mturi, N., Drakeley, C. J., Savarese, B., Villafana, T., Ballou, W. R., Cohen, J., Riley, E. M., Lemnge, M. M., Marsh, K., von Seidlein, L. (2008) Efficacy of RTS,S/AS01E vaccine against malaria in children 5 to 17 months of age. N Engl J Med 359, 2521-32.
  • Bongfen, S. E., Torgler, R., Romero, J. F., Renia, L., Corradin, G. (2007) Plasmodium berghei infected primary hepatocytes process and present the circumsporozoite protein to specific CD8+ T cells in vitro. J Immunol 178, 7054-7063.
  • Carvalho, L. H., Sano, G., Hafalla, J. C., Morrot, A., Curotto de Lafaille, M. A., Zavala, F. (2002) IL-4 secreting CD4+ T cells are crucial to the development of CD8+ T-cell-responses against malaria liver stages. Nat Med 8, 166-170.
  • Epstein J E, Tewari K, Lyke K E, Sim B K, Billingsley P F, Laurens M B, Gunasekera A, Chakravarty S, James E R, Sedegah M, Richman A, Velmurugan S, Reyes S, Li M, Tucker K, Ahumada A, Ruben A J, Li T, Stafford R, Eappen A G, Tamminga C, Bennett J W, Ockenhouse C F, Murphy J R, Komisar J, Thomas N, Loyevsky M, Birkett A, Plowe C V, Loucq C, Edelman R, Richie T L, Seder R A, Hoffman S L (2011) Live attenuated malaria vaccine designed to protect through CD8+ T cell immunity. Science. October 28; 334(6055):475-80. Epub 2011 Sep. 8.
  • Goodman A L, Epp C, Moss D, Holder A A, Wilson J M, Gao G P, Long C A, Remarque E J, Thomas A W, Ammendola V, Colloca S, Dicks M D, Biswas S, Seibel D, van Duivenvoorde L M, Gilbert S C, Hill A V, Draper S J. (2010) New candidate vaccines against blood-stage Plasmodium falciparum malaria: prime-boost immunization regimens incorporating human and simian adenoviral vectors and poxyiral vectors expressing an optimized antigen based on merozoite surface protein 1. Infect Immun. 78(11):4601-4612.
  • Grüner, A. C., Mauduit, M., Tewari, R., Romero, J. F., Depinay N., Kayibanda, M., Lallemand, E., Chavatte, J. M., Crisanti, A., Sinnis, P., Mazier, D., Corradin, G., Snounou, G., Renia, L. (2007) Sterile protection against malaria is independent of immune responses to the circumsporozoite protein. PLoS One 2, e 1371.
  • Haldar, K., Murphy, S. C., Milner, D. A. Jr, Taylor, T. E. (2007) Malaria: mechanisms of erythrocytic infection and pathologic correlates of severe disease. Annu Rev Pathol Mech Dis 2, 217-249.
  • Hoffman, S. L., Goh, L. M. L., Luke, T. C., Schneider, I., Le, T. P., Doolan, D. L., Sacci, J., de la Vega, P., Dowler, M., Paul, C., Gordon, D. M., Stoute, J. A., Church, L. W., Sedegah, M., Heppner, D. G., Ballou, W. R., Richie, T. L. (2002) Protection of humans against malaria with radiation-attenuated Plasmodium falciparum sporozoites. J Infect Dis 185, 1155-64.
  • Jobe, O., Lumsden, J., Mueller, A. K., Williams, J., Silva-Rivera, H., Kappe, S. H., Schwenk, R. J., Matuschewski, K., Krzych, U. (2007) Genetically attenuated Plasmodium berghei liver stages induce sterile protracted protection that is mediated by major histocompatibility complex Class I-dependent interferon-gamma-producing CD8+ T cells. J Infect Dis. 196, 599-607.
  • Klotz, F. W., Scheller, L. F., Seguin, M. C., Kumar, M. A., Marietta, A., Green, S. J., Azad, A. F. (1995) Co-localization of inducible nitric oxide synthase and Plasmodium berghei in hepatocytes from rats immunized with irradiated sporozoites. J Immunol. 154, 3391-3395.
  • Leiriao, P., Mota, M. M., Rodriguez, A. (2005) Apoptoptic Plasmodium-infected hepatocytes provide antigens to liver dendritic cells. J Infect Dis. 191, 1576-1581.
  • Mauduit, M., Tewari, R., Depinay, N., Kayibanda, M., Lallemand, E., Chavatte, J. M., Snounou, G., Renia, L., Grüner, A. C. (2010) Minimal role for the circumsporozoite protein in the induction of sterile immunity by vaccination with live rodent malaria sporozoites. Infect Immun. 78, 2182-8.
  • Mueller, A. K., Labaied, M., Kappe, S. H. & Matuschewski, K. (2005 a) Genetically modified Plasmodium parasites as a protective experimental malaria vaccine. Nature 433, 164-7.
  • Mueller, A. K., Camargo, N., Kaiser, K., Andorfer, C., Frevert, U., Matuschewski, K., Kappe, S. H. (2005 b) Plasmodium liver stage developmental arrest by depletion of a protein at the parasite-host interface. Proc Natl Acad Sci USA 102, 3022-7.
  • Mueller, A. K., Deckert, M., Heiss, K., Goetz, K., Matuschewski, K., Schluter, D. (2007) Genetically attenuated Plasmodium berghei liver stages persist and elicit sterile protection primarily via CD8 T cells. Am J Pathol 171, 107-115.
  • Mwangi, T. W., Ross, A., Snow, R. W., Marsh, K. (2005) Case definitions of clinical malaria under different transmission conditions in Kilifi District, Kenya. J Infect Dis 191, 1932-9.
  • Nussenzweig, R. S., Vanderberg, J., Most, H. & Orton, C. (1967) Protective immunity produced by the injection of γ-irradiated sporozoites of Plasmodium berghei. Nature 216, 160-2.
  • Oliveira, G. A., Kumar, K. A., Calvo-Calle J. M., Othoro, C., Altszuler, D., Nussenzweig, V., Nardin, E. H. (2008) Class II restricted immunity induced by malaria sporozoites. Infect Immun. 76, 200-1206.
  • Prudenco, M., Rodriguez, A., Mota, M. M. (2006) The silent path to thousands of merozoites: the Plasmodium liver stage. Nat Rev Microbiol 4, 849-856.
  • Putrianti, E. D., Silvie, O., Kordes M., Borrmann, S., Matuschewski K. (2009) Vaccine-like immunity against malaria by repeated causal-prophylactic treatment of liver-stage Plasmodium parasites. J Infect Dis 199, 899-903.
  • Riley E. M. (1999) Is T-cell priming required for initiation of pathology in malaria infections? Immunol Today. 20:228-33.
  • Roestenberg, M., McCall, M., Hopman, J., Wiersma, J., Luty, A. J., van Gemert, G. J., van de Vegte-Bolmer, M., van Schaijk, B., Teelen, K., Arens, T., Spaarman, L., de Mast, Q., Roeffen, W., Snounou, G., Renia, L., van der Ven, A., Hermsen, C. C., Sauerwein, R. (2009) Protection against a malaria challenge by sporozoite inoculation. N Engl J Med 361, 468-77.
  • Semblat J P, Silvie O, Franetich J F, Mazier D. (2005) Laser capture microdissection of hepatic stages of the human parasite Plasmodium falciparum for molecular analysis. Methods Mol Biol. 293:301-7.
  • Silvie, O., Semblat, J. P., Franetich, J. F., Hannoun, L., Eling, W., Mazier, D. (2002) Effects of irradiation on Plasmodium falciparum sporozoite hepatic development: implications for the design of pre-erythrocytic malaria vaccines. Parasite Immunol. 24, 221-3.
  • Silvie, O., Goetz, K., Matuschewski, K. (2008) A sporozoite asparagine-rich protein controls initiation of Plasmodium liver stage development. PLoS Pathog. 4(6), e1000086.
  • Sturm, A., Amino, R., van de Sand, C., Regen, T., Retzlaff, S., Rennenberg, A., Krueger, A., Pollok, J. M., Menard, R., Heussler, V. T. (2006) Manipulation of host hepatocytes by the malaria parasite for delivery into liver sinusoids. Science 313, 1287-1290.
  • Tarun, A. S., Dumpit, R. F., Camargo, N., Labaied, M., Liu, P., Takagi, A., Wang, R., Kappe, S. H. (2007) Protracted sterile protection with Plasmodium yoelii pre-erythrocytic genetically attenuated parasite malaria vaccines is independent of significant liver stage persistence and is mediated by CD8+ T cells. J Infect Dis 196, 608-16.
  • van Dijk, M. R., Douradinha, B., Franke-Fayard, B., Heussler, V., van Dooren, M. W., van Schaijk, B., van Gemert, G. J., Sauerwein, R. W., Mota, M. M., Waters, A. P., Janse, C. J. (2005) Genetically attenuated, P36p-deficient malarial sporozoites induce protective immunity and apoptosis of infected liver cells. Proc Natl Acad Sci USA. 102, 12194-9.
  • van Schaijk, B. C., Janse, C. J., van Gemert, G. J., van Dijk, M. R., Gego, A., Franetich, J. F., van de Vegte-Bolmer, M., Yalaoui, S., Silvie, O., Hoffman, S. L., Waters, A. P., Mazier, D., Sauerwein, R. W., Khan, S. M. (2008) Gene disruption of Plasmodium falciparum p52 results in attenuation of malaria liver stage development in cultured primary hepatocytes. PLoS ONE. 3:e3549.
  • VanBuskirk, K. M., O'Neill, M. T., De La Vega, P., Maier, A. G., Krzych, U., Williams, J., Dowler, M. G., Sacci, J. B. Jr., Kangwanrangsan, N., Tsuboi, T., Kneteman, N. M., Heppner, D. G. Jr., Murdock, B. A., Mikolajczak, S. A., Aly, S. A., Cowman, A. F., Kappe, S. H. (2009) Preerythrocytic, live-attenuated Plasmodium falciparum vaccine candidates by design. Proc Natl Acad Sci USA. 106, 13004-9.
  • White, K. L., Synder, H. L., Krzych, U. (1996) MHC class-I dependent presentation of exoerythrocytic antigens to CD8+ T lymphocytes is required for protective immunity against Plasmodium berghei. J Immunol. 156, 3374-3381.

Claims

1. A composition comprising at least two polypeptides, or one or more nucleic acid molecules encoding at least two polypeptides, wherein each of the polypeptides comprises an amino acid sequence selected from MAL13P1.13 (SEQ ID NO. 1), PF14_0480 (SEQ ID NO. 2), MAL13P1.258 (SEQ ID NO. 3), PF14_0435 (SEQ ID NO. 4), PF14_0390 (SEQ ID NO. 5), PF13_0242 (SEQ ID NO. 6), PF13_0168 (SEQ ID NO. 7), PF11_0342/0421 (SEQ ID NO. 8), PFc0515c (SEQ ID NO. 9), PFc0135c (SEQ ID NO. 10), PF14_0533 (SEQ ID NO. 11), PF13_0282 (SEQ ID NO. 12), PF10_0081 (SEQ ID NO. 13), PFF0420c (SEQ ID NO. 14), PF14_0323 (SEQ ID NO. 15), PF10_0375 (SEQ ID NO. 16), PF11_0128 (SEQ ID NO. 17), PF14_0548 (SEQ ID NO. 18), PF08_0051 (SEQ ID NO. 19), PF08_0036 (SEQ ID NO. 20), PF13_0135 (SEQ ID NO. 21), PF13_0343 (SEQ ID NO. 22), PFE1260c (SEQ ID NO. 23), fragments of the above amino acid sequences.

amino acid sequences that are at least 80% identical to any of the above amino acid sequences, and

2. (canceled)

3. The composition of claim 1, comprising further polypeptide(s), wherein the one or more further polypeptide(s) comprise an amino acid sequence selected from PF14_0530 (SEQ ID NO. 24), Mal8P1.134 (SEQ ID NO. 25), amino acid sequences that have at least 80% sequence identity to any of the amino acid sequences of SEQ ID NOs. 24 and 25, and fragments of the above amino acid sequences.

4. The composition of claim 1, wherein the at least two polypeptides are selected from polypeptides comprising an amino acid sequence of SEQ ID NOs. 1 to 25 or fragment(s) thereof or a fragment of one of said amino acid sequences.

5. The composition of claim 1, comprising at least three, of the polypeptides or fragments of the polypeptides.

6. (canceled)

7. The composition of claim 1, comprising at least two polypeptides each comprising an amino acid sequence selected from MAL13P1.13 (SEQ ID NO. 1), PF14_0480 (SEQ ID NO. 2), MAL13P1.258 (SEQ ID NO. 3), PF14_0435 (SEQ ID NO. 4), PF14_0530 (SEQ ID NO. 24), PF14_0323 (SEQ ID NO. 15), amino acid sequences having at least 80% sequence identity to any of the amino acid sequences of SEQ ID NOs. 1 to 4, 15 and 24, and fragments thereof.

8. The composition of claim 1, wherein the fragment comprises at least one antigenic determinant or epitope of the amino acid sequence.

9. The composition of claim 8, wherein the antigenic determinant or epitope is a CD8+ T cell epitope, a CD4+ T cell epitope.

10. The composition of claim 8, wherein the fragment comprises at least one amino acid sequence selected from (SEQ ID NO. 26) SLICGLYLL, (SEQ ID NO. 27) ILYSLMINSL, (SEQ ID NO. 28) LICGLYLLTL, (SEQ ID NO. 29) VLLEKINVI, (SEQ ID NO. 30) YLSPNFINKI, (SEQ ID NO. 31) ILHGGVYRL, (SEQ ID NO. 32) ILFLFILSI, (SEQ ID NO. 33) LLFINEINKL, (SEQ ID NO. 34) SLISLYIYYV, (SEQ ID NO. 35) FLLLMLVSI, (SEQ ID NO. 36) FLTLMARKL, (SEQ ID NO. 37) NLLDPLVVV, (SEQ ID NO. 38) LLLEGNFYL, (SEQ ID NO. 39) KLIPVNYEL, and[[/or]] (SEQ ID NO. 40) ILIPSLPLI.

11. The composition of claim 10, comprising at least one fragment comprising an amino acid sequence selected from

SEQ ID NOs. 26 to 28,
SEQ ID NOs. 29 and 30,
SEQ ID NOs. 31 to 33,
SEQ ID NOs. 34 and 35,
SEQ ID NO. 36, and
SEQ ID NOs. 37 to 40.

12. The composition according to claim 1, wherein the polypeptides or fragments thereof comprise one or more labels, N- and/or C-terminal modifications, drug(s) or other agents.

13. The composition of claim 1 comprising at least two nucleic acid molecules each encoding at least one polypeptide, or fragment thereof, according to claim 1.

14. The composition of claim 13, comprising at least three of the nucleic acid molecules.

15. The composition according to claim 1, further comprising a carrier and/or an adjuvant.

16. The composition of claim 15, wherein the carrier is a virus particle or a part thereof, an envelope protein of a viral vector or of a virus particle, or a nanocarrier.

17. The composition of claim 16, wherein the virus particle is a Hepatitis 13 virus particle or a part thereof.

18. The composition of claim 16, wherein the nanocarrier is a cell-targeted nanocarrier.

19. The composition of claim 15, wherein the adjuvant triggers a CD8 T cell response.

20. A multicomponent malaria vaccine, which comprises a composition of claim 1.

21. A method for vaccinating against malaria, wherein said method comprises administering, to a subject in need of such vaccination, a polypeptide, or a nucleic acid molecule encoding said polypeptide, wherein the polypeptide comprises an amino acid sequence selected from SEQ ID NOs. 1 to 23 or an amino acid sequence that is at least 80% identical to any of said amino acid sequences,

or a fragment thereof.

22. The method, according to claim 21, wherein the polypeptide comprises an amino acid sequence selected from SEQ ID NOs. 26 to 36.

23. The method, according to claim 21, comprising administering to the subject a nucleic acid molecule encoding at least one polypeptide or fragment thereof according to claim 21.

24. The method, according to claim 23, wherein said nucleic acid molecule has a nucleotide sequence selected from SEQ ID NOs. 41 to 63.

25. The composition of claim 1, comprising at least five of the polypeptides or fragments of the polypeptides.

26. The composition of claim 13, comprising at least five of the nucleic acid molecules.

Patent History
Publication number: 20130323276
Type: Application
Filed: Dec 6, 2011
Publication Date: Dec 5, 2013
Applicant: RUPRECHT-KARLS-UNIVERSITÄT HEIDELBERG (HEIDELBERG)
Inventors: Johannes Pfeil (Heidelberg), Kirsten Heiss (Heidelberg), Ann-Kristin Müller (Dossenheim)
Application Number: 13/988,339