• Tidak ada hasil yang ditemukan

genetically modified organisms in containment

N/A
N/A
Protected

Academic year: 2023

Membagikan "genetically modified organisms in containment "

Copied!
15
0
0

Teks penuh

(1)

Send to Environmental Protection Authority preferably by email (neworganisms@epa.govt.nz) or alternatively by post (Private Bag 63002, Wellington 6140)

Payment must accompany final application; see our fees and charges schedule for details.

To obtain approval for projects to develop

genetically modified organisms in containment

Application Number

APP204179

Date

05-May-2021

(2)

Completing this application form

1. This form has been approved under section 42A of the Hazardous Substances and New

Organisms (HSNO) Act 1996. It only covers projects for development (production, fermentation or regeneration) of genetically modified organisms in containment. This application form may be used to seek approvals for a range of new organisms, if the organisms are part of a defined project and meet the criteria for low risk modifications. Low risk genetic modification is defined in the HSNO (Low Risk Genetic Modification) Regulations:

http://www.legislation.govt.nz/regulation/public/2003/0152/latest/DLM195215.html.

2. If you wish to make an application for another type of approval or for another use (such as an emergency, special emergency or release), a different form will have to be used. All forms are available on our website.

3. It is recommended that you contact an Advisor at the Environmental Protection Authority (EPA) as early in the application process as possible. An Advisor can assist you with any questions you have during the preparation of your application.

4. Unless otherwise indicated, all sections of this form must be completed for the application to be formally received and assessed. If a section is not relevant to your application, please provide a comprehensive explanation why this does not apply. If you choose not to provide the specific information, you will need to apply for a waiver under section 59(3)(a)(ii) of the HSNO Act. This can be done by completing the section on the last page of this form.

5. Any extra material that does not fit in the application form must be clearly labelled, cross- referenced, and included with the application form when it is submitted.

6. Please add extra rows/tables where needed.

7. You must sign the final form (the EPA will accept electronically signed forms) and pay the application fee (including GST) unless you are already an approved EPA customer. To be recognised by the EPA as an “approved customer”, you must have submitted more than one application per month over the preceding six months, and have no history of delay in making payments, at the time of presenting an application.

8. Information about application fees is available on the EPA website.

9. All application communications from the EPA will be provided electronically, unless you specifically request otherwise.

Commercially sensitive information

10. Commercially sensitive information must be included in an appendix to this form and be identified as confidential. If you consider any information to be commercially sensitive, please show this in the relevant section of this form and cross reference to where that information is located in the confidential appendix.

11. Any information you supply to the EPA prior to formal lodgement of your application will not be publicly released. Following formal lodgement of your application any information in the body of this application form and any non-confidential appendices will become publicly available.

(3)

12. Once you have formally lodged your application with the EPA, any information you have supplied to the EPA about your application is subject to the Official Information Act 1982 (OIA). If a request is made for the release of information that you consider to be confidential, your view will be considered in a manner consistent with the OIA and with section 57 of the HSNO Act. You may be required to provide further justification for your claim of confidentiality.

Definitions

Containment

Restricting an organism or substance to a secure location or facility to prevent escape. In respect to genetically modified organisms, this includes field testing and large scale fermentation

Controls

Any obligation or restrictions imposed on any new organism, or any person in relation to any new organism, by the HSNO Act or any other Act or any regulations, rules, codes, or other documents made in accordance with the provisions of the HSNO Act or any other Act for the purposes of controlling the adverse effects of that organism on people or the environment

Genetically Modified Organism (GMO)

Any organism in which any of the genes or other genetic material:

• Have been modified by in vitro techniques, or

• Are inherited or otherwise derived, through any number of replications, from any genes or other genetic material which has been modified by in vitro techniques

New Organism

A new organism is an organism that is any of the following:

• An organism belonging to a species that was not present in New Zealand immediately before 29 July 1998;

• An organism belonging to a species, subspecies, infrasubspecies, variety, strain, or cultivar prescribed as a risk species, where that organism was not present in New Zealand at the time of promulgation of the relevant

regulation;

• An organism for which a containment approval has been given under the HSNO Act;

• An organism for which a conditional release approval has been given under the HSNO Act;

• A qualifying organism approved for release with controls under the HSNO Act;

• A genetically modified organism;

• An organism belonging to a species, subspecies, infrasubspecies, variety, strain, or cultivar that has been eradicated from New Zealand;

• An organism present in New Zealand before 29 July 1998 in contravention of the Animals Act 1967 or the Plants Act 1970. This does not apply to the organism known as rabbit haemorrhagic disease virus, or rabbit calicivirus A new organism does not cease to be a new organism because:

• It is subject to a conditional release approval; or

• It is a qualifying organism approved for release with controls; or

• It is an incidentally imported new organism

Project An individual or collaborative endeavour that is planned to achieve a particular aim or research goal

(4)

1. Applicant details

1.1. Applicant

Company Name: (if applicable) Ruakura Technologies Limited (RTL) Contact Name: Steve Hodgkinson

Job Title: Chief Executive Officer

Physical Address: Ruakura Research Centre, 10 Bisley Rd, Enderley, Hamilton 3214

Postal Address

(provide only if not the same as the physical)

:

10 Bisley Rd, Private Bag, 3123, Hamilton 3214.

Phone (office and/or mobile): 021 995 389 Fax: N/A

Email: steve.hodgkinson@ruatech.co.nz

1.2. New Zealand agent or consultant (if applicable)

Company Name:

Contact Name:

Job Title:

Physical Address:

Postal Address (provide only if not the same as the physical): Phone (office and/or mobile):

Fax:

Email:

(5)

2. Information about the application

2.1. Brief application description

Approximately 30 words about what you are applying to do

To develop low-risk genetically modified microorganisms and mammalian cells. The resultant modified organisms will be used to generate protein for purification and enrichment.

2.2. Summary of application

Provide a plain English, non-technical description of what you are applying to do and why you want to do it

Ruakura Technologies Limited focuses on specialist technical service provision and

product development. We seek permission to develop and work with genetically modified organisms (GMO) that produce specific and targeted proteins or partial proteins that are of interest for subsequent purification. These targets will be used in biological

applications, primarily for use as antigens for the generation of antibodies in animal- based production systems. Proteins of interest will be selected based on theoretical knowledge and suitability for use as an antigen; for example, viral or bacterial proteins presenting external surfaces would provide strong candidates. RTL would exclude any protein targets known to be toxic or damaging to humans or animals. In addition, no live pathogens will be used for this project, nor will any of the genetic targets have the ability to produce live pathogens.

2.3. Technical description

Briefly describe the host organism(s) and the proposed genetic modifications. Please make sure that any technical words used are included in a glossary. Note if any part of this research project is already covered by an existing HSNO Act approval that your organisation holds or uses.

RTL proposes to develop GMOs from commercially available low-risk microorganisms such as non-pathogenic Escherichia coli and immortalised mammalian cells; Cricetulus griseus (Chinese Hampster), Homo sapiens (Human) and Mus Musculus (Mouse). All organisms are Category 1 organisms as defined in section 7 of the HSNO (Low-Risk Genetic Modification) regulations 2003.

Modification of organisms would incorporate genetic elements in the form of an expression vector, which contains the genetic information to allow expression of the protein of interest (i.e. promoter, gene, secretion signal and terminator), as well as a

(6)

selection cassette (i.e. promoter, antibiotic selection cassette and terminator) for the generation of stable cell lines via integration into the host genome, the vector would also contain other standard elements as outlined in 4.1 below. Expression vectors would be introduced using standard molecular techniques, including heat shock, transfection and/or electroporation, allowing for the expression of the proteins of interest. Both transient (i.e. temporary expression) and stable cell lines (i.e. DNA stably integrated into the host cell) will be generated to allow purification of the proteins of interest. In the first instance, transient expression (up to 10 days following transfection) would be used to screen and to inform which protein/s of interest are the best suited for the subsequent generation of stable cell lines. This would be done primarily by quantification of the amount of secreted protein. Transient cultures will be destroyed following a culture period long enough to allow the production of the protein of interest, while stable cell lines would be banked (cryopreservation in LN2) for reuse as required. An example of a commercially available system (likely to be, or, very similar to that which would be for the proposed work) for producing protein in mammalian cells is referenced below (1-2) and well as a reference demonstrating recombinant protein expression using this system (3). The resultant modified organisms would be screened to allow selection of those organisms best producing the target protein of interest. Organisms would then be expanded in culture to enable further production and purification of these targets to levels suitable for use as an antigen (milligram to gram amounts). All culture of both bacterial and mammalian GMOs would be at small to medium scale (i.e. <10L to a maximum of 25L) scale.

3. Information about the new organism(s)

3.1. Identity of the host organism(s) For each host organism:

• Provide its taxonomic name and describe what type of organism it is.

• Provide a description of the strain(s) being applied for (if relevant).

• If the host organism is derived from humans (eg, cell lines) or may have cultural significance (e.g.

sourced from native biota), provide details of its source.

• State the category (Category 1 or Category 2) of the host organism (as per the HSNO (Low Risk Genetic Modification) Regulations).

(7)

Organism Taxonomic name

Organism Category

/GMO Category*

Source, Comment/Reference.

Bacteria Escherichia coli, Migula 1895 Castellani and Chalmers 1919.

1/A Comercially available Non-pathogenic laboratory strains (e.g ATCC:12435, K12 or derivative).

Mammalian Chinese hamster

Cricetulus griseus, Milne-Edwards 1867

CHO K1

1/A Comerically available cell lines (e.g. ATCC:

CCL-61, CHO), Kao F. T., Puck T. T.

(1968).

Mammalian

Human Homo sapiens, sub species sapien, Human Linnaeus 1758

1/A Comercially available cell lines (e.g. ATCC:

CRL-1573, HEK293 and derivatives), Thomas., Smart (2005).

Mammalian

Mouse Mus Musculus, Mouse, Linnaeus 1758

1/A Commercially available cell lines (e.g.

ATCC: CCL-131, Neuro2A), Olmsted J.B.

(1970).

* Host Organism and GMO Category as defined in the HSNO (low-Risk Genetic modification) regulations 2003.

3.2. Regulatory status of the organism

Is the organism that is the subject of this application also the subject of:

An innovative medicine application as defined in section 23A of the Medicines Act 1981?

Yes

No

An innovative agricultural compound application as defined in Part 6 of the Agricultural Compounds and Veterinary Medicines Act 1997?

Yes

No

4. Information about the project

4.1. Describe the nature and range of the proposed genetic modifications

• Describe the nature and range of the proposed genetic modifications (e.g. the range of elements that the vectors or gene constructs may contain, and the type, source and function of the donor genetic material).

(8)

• State the category (Category A or Category B) of the genetic modifications (as per the HSNO (Low Risk Genetic Modification) Regulations).

For modifications, RTL will use standard, non-conjugative, cloning and expression vectors to genetically modify host cells. Genetic sequences of the proteins, fusion proteins or partial proteins of interest will be synthetically generated (commercial supplier) and cloned into expression plasmids for transfection and modification of E. coli. Subsequent growth of E. coli allows sufficient quantities of the expression vector to be produced, which would then be isolated/purified and used to modify ensuing and approved microbial or mammalian host organisms to produce the protein or partial protein of interest. Transformation of E.coli would be performed using standard molecular

techniques of heat shock or electroporation. Transfection of mammalian cells would be achieved using established methods of transfection, including commercially available lipid-based transfection reagents (for example, lipofectamine and expifectamine) with electroporation providing an alternative means of transfecting target DNA. Synthetic material for the expression of individual proteins will be derived primarily from microbial sequences as obtained from databases such as the National Centre for Biotechnology Information, only DNA sequences coding for known proteins will be used to generate synthetic DNA. Sequences derived from pathogenic organisms will be used but, no novel sequences from pathogenic organisms or sequences that code for proteins known to be toxic will be used. It is not expected that any of the proteins produced would have vertebrate toxicity with an LD50 of less than 100µg/kg. In fact, toxic proteins are extremely unlikely to be able to be made in a mammalian expression system, as proposed for this work, as the production of toxic proteins would be detrimental to the culture of the mammalian cells and likely to render the cultures unviable. Expression vectors may contain promoters and regulatory elements, transcriptional terminator signals, inducible expression elements, reporter genes, selectable marker genes,

secretory and targeting signals, recombination sites, a microbial or viral surface whole or partial gene, recombination sites, flanking sequences, insulator sequences, origins of replication, antibiotic selection genes and sequences that facilitate recombination, protein solubility, protein stability and protein purification.

All modifications will be Category A modifications as defined in the HSNO (low-Risk Genetic modification) regulations 2003.

Modifications to cell lines will be restricted to modifications that do not lead to the production of infectious particles such as prions or replication-competent viruses and do not increase the risk of the cell line to humans. Cell culture will be carried out under PC1 containment as described in the appended AgResearch Ruakura Transitional and

Containment Facility Manual. The cell lines to be used require specific culture conditions for survival and would not survive in the natural environment outside of the laboratory,

(9)

As such, they do not pose a risk of escape into the environment. Therefore, the nature of containment is such that the risk of escape and subsequent infection is negligible.

4.2. Proposed containment of the new organism(s) (physical and operational)

• State which Containment Standard(s) your facility is approved to.

• State the minimum containment level (PC1 or PC2 as per AS/NZS2243.3:2002) required to contain the GMOs (as per the HSNO (Low Risk Genetic Modification) Regulations).

• Discuss whether controls in addition to the requirements listed in the Standard(s) are necessary to adequately contain the GMOs.

Ruakura Technologies Ltd is physically located within the AgResearch-Ruakura Campus (MPI approved containment facilities, premises number 2501). RTL is an authorised user of these facilities, and they are approved to standards: 154.02.17 (Transitional Facilities for Biological Products) and 154.03.02 (Facilities for Microorganisms and Cell Culture).

These facilities are designed and constructed to contain the approved organisms held within them under reasonably foreseeable circumstances. These facilities will be operated in accordance with the AgResearch Ruakura Campus Containment Facility Manual. These facilities comprise containment laboratory areas, which meet regulatory requirements and are inspected regularly. A set of controls has been proposed, and a management plan based on the approval controls is included in the Containment Manual.

Work involving Risk Group 1 microorganisms and animal cell lines will be carried out under PC1 containment as described in the AgResearch Ruakura Transitional and

Containment Facility Manual. Access is restricted to appropriately trained and authorised personnel.

For up-scaled culture of microorganisms or mammalian cells, dual containment would be standard, the first level being the culture system itself. Fermentation would be the method of choice for scaled bacterial culture with a maximum volume of 10L. The fermentation vessel provides primary containment, and the fermenter, whilst running, would be housed in a secondary container with sufficient capacity to catch all culture material in the unlikely event of a fermenter failure. For mammalian cells, a similar approach would be used using a portable wave bag system. These systems use liquid filled multi skinned bladders and gentle rocking to culture cells. As with fermentation, secondary containment would be employed in case of primary vessel failure. All waste material would be handled following prescribed decontamination protocols as described in the AgResearch containment manual. In addition, specialised spill kits will be located in close proximity to all cultures to enable an immediate response in the unlikely event of a vessel failure.

(10)

Experimental work to develop and use cell lines would be carried out in containment laboratories at the Ruakura Research Campus under conditions designed to minimise the risk of infection of personnel. Genetically modified cell lines will not be applied to animals or humans.

Should this application be approved, the work performed by RTL will be governed and administered by the Ruakura Transitional Containment Facility. Individual RTL

researchers using this approval will ensure that the project meets the criteria for low-risk research described in the application; the new organism they wish to work with matches the organism description in the approval; the research is consistent with the purpose of the application; and that there are no other risk factors. If an individual researcher’s project does not meet these requirements, RTL will need to apply to the EPA, as appropriate, for a new ‘development in containment’ approval.

(11)

5. Risks, costs and benefits

Provide information of the risks, costs and benefits of the GMOs in the following areas of impact:

• The environment.

• Human health and safety.

• The economy (e.g. the ability of people and communities to provide for their economic wellbeing).

• The relationship of Māori and their culture and traditions with their ancestral lands, water, sites, waahi tapu, valued flora and fauna and other taonga, and the principles of the Treaty of Waitangi (The details of any engagement or consultations with Māori that you have undertaken in relation to this application should be discussed here).

• Society and community.

• New Zealand’s international obligations.

All organisms RTL seeks approval to modify are classed as category 1 organisms, as defined in the HSNO (low-Risk Genetic modification) regulations 2003, modifications are category A as defined in the Act. None of the proposed modifications would be expected to introduce traits that are in any way likely to increase pathogenicity, virulence or the infectivity of host species and are not in any way expected to adversely affect humans, the community, animals, plants or microbes.

All cell lines derived from animals or humans are entirely reliant on human intervention to survive and are therefore incapable of survival outside the laboratory environment and pose negligible risk to laboratory personal. The nature of the containment is such that there is a negligible risk with respect to escape, people, animals, and the environment.

In the unlikely event of an incident or accident that may lead to a breach in containment, contingency plans have been developed and are described in the AgResearch Ruakura Transitional and Containment Facility Manual. This includes well-articulated instructions should a breach occur, how this is to be managed and who should be notified.

Staff will be trained on the risk involved in working with the host organisms defined above and on good laboratory practice to ensure adherence to all containment measures.

If approved, the generation of GMOs as outlined in this application is likely to have the benefit of creating jobs as the work programme of RTL grows.

RTL, primarily through our Chairman (Waikato, Tainui, Taranaki, Te Arawa), has carried the mantle of providing leadership with regards to our work, especially noting an

association with tangata whenua wherever we have been looking at potential projects.

He has facilitated introductions to those with mana whenua status who have then

provided further direction for us. It has been remarkable to note that since we have been working with the availability of such guidance, our confidence and capability of inclusion from a Maori perspective continues to grow.

(12)

The tangata whenua status of our Chairman at Ruakura, has proven valuable for our engagement with several Maori trusts concerning this work. We have had discussions with the lead member of the Authors of “Te Mata Ira”, the “Guidelines for Genomic Research with Maori” (Oct 2016) from the University of Waikato. Therefore, it is not surprising that as a result of the steerage we have been receiving, no objections have been noted to date, and the GMO modification work RTL seeks approval for does not involve valued native flora, fauna or other taonga.

6. Other information

Add here any further information you wish to include in this application including if there are any ethical considerations that you are aware of in relation to your application.

N/A

(13)

7. Checklist

This checklist is to be completed by the applicant

Application Comments/justifications

All sections of the application form completed or you have requested an information waiver under section 59 of the HSNO Act

Yes

No

(If No, please discuss with an Advisor to enable your application to be further processed)

N/A

Confidential data as part of a separate, identified appendix

Yes

No

Supplementary optional information attached:

• Copies of additional references

Yes

No

• Relevant correspondence

Yes

No Administration

Are you an approved EPA customer?

Yes

No If Yes are you an:

Applicant:

Agent:

RTL is a newly formed company and has not had any reason to engage with the EPA before now.

If you are not an approved customer, payment of fee will be by:

• Direct credit made to the EPA bank account (preferred method of payment) Date of direct credit:

• Cheque for application fee enclosed

Yes

No

Payment to follow

Yes

No

Payment to follow

Electronic, signed copy of application e- mailed to the EPA

Yes

(14)

Signature of applicant or person authorised to sign on behalf of applicant

I am making this application, or am authorised to sign on behalf of the applicant or applicant organisation.

I have completed this application to the best of my ability and, as far as I am aware, the information I have provided in this application form is correct.

05-May-2021 Signature Date

Request for information waiver under section 59 of the HSNO Act

I request for the Authority to waive any legislative information requirements (i.e. concerning the information that has been supplied in my application) that my application does not meet (tick if applicable).

Please list below which section(s) of this form are relevant to the information waiver request:

N/A

(15)

Appendices and referenced material (if any) and glossary (if required)

References:

1. Expi293 Expression system user guide.

User Guide: Expi293 Expression System (thermofisher.com) 2. Vector map pcDNA 3.4-TOPO.

pcdna3_4_topo_ta_map.pdf (thermofisher.com)

3. Efficient and inexpensive transient expression of multispecific multivalent antibodies in Expi293 cells. Fang et al 2017.

(PDF) Efficient and inexpensive transient expression of multispecific multivalent

antibodies in Expi293 cells (researchgate.net)

Referensi

Dokumen terkait

Penulis pada bab 3 ini akan membahas analisis terkait peran pemerintah Nigeria dalam menjaga stabilitas keamanan terhadap gerakan teroris Boko Haram pada tahun 2010-2016