WO2022031992A1 - Lysis buffer compositions and methods for preparing a viral biological sample useful for covid-19 testing - Google Patents

Lysis buffer compositions and methods for preparing a viral biological sample useful for covid-19 testing Download PDF

Info

Publication number
WO2022031992A1
WO2022031992A1 PCT/US2021/044781 US2021044781W WO2022031992A1 WO 2022031992 A1 WO2022031992 A1 WO 2022031992A1 US 2021044781 W US2021044781 W US 2021044781W WO 2022031992 A1 WO2022031992 A1 WO 2022031992A1
Authority
WO
WIPO (PCT)
Prior art keywords
lysis buffer
rna
sample
biological sample
virus
Prior art date
Application number
PCT/US2021/044781
Other languages
French (fr)
Inventor
Klaus HOFFMEIER
Johannes Dapprich
Karl P. DRESDNER
Bjorn ROTTER
Original Assignee
Rely Biotech Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Rely Biotech Inc. filed Critical Rely Biotech Inc.
Publication of WO2022031992A1 publication Critical patent/WO2022031992A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1003Extracting or separating nucleic acids from biological samples, e.g. pure separation or isolation methods; Conditions, buffers or apparatuses therefor
    • C12N15/1006Extracting or separating nucleic acids from biological samples, e.g. pure separation or isolation methods; Conditions, buffers or apparatuses therefor by means of a solid support carrier, e.g. particles, polymers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1003Extracting or separating nucleic acids from biological samples, e.g. pure separation or isolation methods; Conditions, buffers or apparatuses therefor
    • C12N15/1006Extracting or separating nucleic acids from biological samples, e.g. pure separation or isolation methods; Conditions, buffers or apparatuses therefor by means of a solid support carrier, e.g. particles, polymers
    • C12N15/1013Extracting or separating nucleic acids from biological samples, e.g. pure separation or isolation methods; Conditions, buffers or apparatuses therefor by means of a solid support carrier, e.g. particles, polymers by using magnetic beads
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6806Preparing nucleic acids for analysis, e.g. for polymerase chain reaction [PCR] assay
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/70Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving virus or bacteriophage
    • C12Q1/701Specific hybridization probes
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to methods and lysis buffer compositions useful for preparing a sample specimen, such as a viral biological sample, for refrigeration-free shipping, storage, and more sensitive, accurate, rapid, safe, SARS-CoV-2 virus testing.
  • SARS-CoV-2 stands for “severe acute respiratory syndrome coronavirus 2”, the virus responsible for causing the coronavirus disease “COVID-19”.
  • SARS- CoV-2 was previously known as “2019 novel coronavirus”.
  • Many documents, information sites, guidelines and testing protocols use the two terms SARS-CoV-2 and COVID-19 somewhat interchangeably but typically refer to testing procedures for the presence of SARS-CoV-2 in individuals that may develop COVID-19 symptoms.
  • BACKGROUND OF THE INVENTION [0003] The COVID-19 virus pandemic which has infected more than 16 million individuals and causing more than 650,000 fatalities since its 2019 outbreak.
  • SARS-CoV-2 virus SARS coronavirus
  • SARS coronavirus infection sites include lower respiratory tract infections, pneumonia, and fatal acute respiratory distress syndrome (ARDS) and infections in other organs.
  • ARDS acute respiratory distress syndrome
  • SARS-CoV-2 infection causes inflammatory diseases which worsen in patients already having lung diseases such as asthma, chronic obstructive pulmonary disease, or emphysema and such patients often need intensive medical care. Elderly and immune diseased persons are at higher risk that young adults.
  • e-gene specific primers of the WHO real-time PCR assay have been reported to generate up to 10% false positives, which have been found to result at least in part from a partial match of the e-gene specific primers intended to identify SARS-CoV-2 to a 70 bp long region that matches a bacterial species common in the nasopharyngeal environment, Neisseria subflava.
  • SARS-CoV-2 test methods have at least one unneeded dilution step. and need better safety measures in place. SARS-CoV-2 test methods need to be less sensitive to ambient temperatures during shipping and storage.
  • SARS-CoV-2 test methods are inconvenient in that they require refrigeration of the sample specimen to protect test accuracy and sensitivity.
  • SARS-CoV-2 test methods are not efficient and rapidly deployed and tested, which causes shocking public realization that there are still significant backlogs, uncertainties and inadequacies in testing for SARS- CoV-2 infection in humans. Delays in SARS-CoV-2 testing of up to two weeks have been reported and to some extent are due to using test methods, which are methodologically defective in their procedural expediency. Even more problematic is that delaying the testing of a biological sample for traces of nucleic acids is fraught with false negative test results.
  • SARS-CoV-2 tests may have faulty designs or incorrectly adapted biological sample preparation methods allowing for significant biological sample deterioration and release of ribonucleases and deoxy-ribonucleases from cells in the biological sample, which causes rapid and significant degradation of the nucleic acids sequences of the SARS-CoV-2 virus in the biological sample needed to accurately detect if the biological specimen from a human shows that the human is infected with SARS-CoV-2 virus.
  • VTM Viral Transport Medium
  • VTM Viral Transport Medium
  • UTM Universal Transport Medium
  • Collection and testing protocols for biological samples often include the need to refrigerate the samples.
  • a major SARS-CoV-2 virus testing lab company, Quest Diagnostics, issued guidelines on June 29, 2020 which include refrigerating biological samples in dry ice that are made ready for transporting and storage prior to their testing.
  • a refrigeration step is not practical for sample collections of the biological matter, such as in the case of virus detection testing of humans living under poor conditions, in wilderness areas, or countries with limited capabilities and resources. It is an unresolved problem in the that human biological samples must be frozen during their transport to centers where the samples can be tested for the presence of SARS- CoV-2 virus test.
  • test kits used on SARS-CoV-2 virus samples need to be accurate despite the fact that the original SARS-CoV-2 virus genome has been found capable of appearing with thousands of random mutations. There are doubts that these SARS-CoV-2 virus test kits are capable of being sufficiently accurate given the number of already known and newly emerging mutations.
  • the SARS-CoV-2 mutation known as D614G mutant may greatly matter for infectivity and disease progression (Schraer, 7- 19-2020 BBC News “Coronavirus: Are mutations making it more infectious?”).
  • D614G mutated SARS-CoV-2 virus has a virus spike protein change which increases infectivity of SARS-CoV-2 virus to humans. Needed are test kits which can accurately detect D614G and other mutations of the SARS-CoV-2 genome to stay aware of the pandemic evolution of SARS-CoV-2 virus genome. [0016] A dependence of SARS-CoV2 infectivity and disease progression to a person’s HLA and MHC type has been observed (Is there a genetic link to COVID-19 disease severity? Free T., BioTechniques, July 27, 2020).
  • COVID- 19 Gene-based risks related to COVID- 19 overlay the currently known risks such as age and pre-existing conditions and are governed primarily by a region on chromosome 6 called HLA/MHC. This region is responsible for the immune system’s defense against pathogens. Evolution has made this region extremely diverse so that - as a species - humans do not get wiped out by a newly emerged pathogen such as the one presented by the current pandemic. Individuals, however, can have very different genetic risk profiles. Studies of COVID- 19 urgently look for information on gene-based risk of infection, disease progression and risk of death. Understanding the correlations among these factors can lead to personalized interventions that can help better protect vulnerable groups and save patients’ lives through specific treatments.
  • Biological samples from many biological matter nucleic acid sources such as a person, will comprise biological fluids, cells, cell molecules, and various enzymes, in addition to the biological matter nucleic acid source infectious containing pathogens such as a virus.
  • the nuclease enzymes in a biological sample can rapidly destroy the RNA or DNA in the biological sample after this sample is obtained and before the testing to detect the RNA or DNA is implemented. There are nuclease enzymes in nearly all biological samples.
  • Nuclease enzymes can hydrolyze nucleic acid phosphodiester bonds and can chemically alter nucleic acid bases. Ribonucleases specifically degrade RNA. Deoxyribonucleases specifically degrade DNA. Non-specific nucleases degrade both RNA and DNA. Polynucleotide phosphorylase and pyrophosphorylase, are also capable of depolymerizing RNA. [0019] Phosphomonoesterases bind to polynucleotides or oligonucleotides at a terminal phosphate group or mononucleotide to remove a phosphate group.
  • Endonucleases produce oligonucleotides which thereby cause a lowering of the viscosity of the nucleic acid as well as decreasing the ability of a test kit to identify the nucleic acid virus.
  • Exonucleases release mononucleotides from nucleic acids. Some nucleases are both endonucleases and exonucleases.
  • the third feature is mode of phosphodiester bond cleavage. Polynucleotides can be cleaved at the phosphodiester bond on either side of the phosphate.
  • nucleases Of the ribonucleases, those enzymes that produce 3' -phosphates and 5' –hydroxyl termini are concerned with the degradation of RNA whereas those enzymes that are involved in the processing of RNA precursor molecules generate 5' - phosphate and 3' -hydroxyl termini. Other distinguishing features of nucleases can be made based on their specificity towards secondary structures, directions of attack of the polynucleotide, and maximum enzymatic nuclease velocity.
  • nucleic acids may be modified by telomerase, also called terminal transferase, a ribonucleoprotein that adds a species-dependent telomere repeat sequence to the 3' end of a telomere, which is a region of repetitive sequences at each end of eukaryotic chromosomes (in most eukaryotes) to protect the end of the chromosome from DNA damage or from fusion with neighboring chromosomes.
  • nucleic acids may be modified by a transposable element (transposon, or jumping gene), which is a DNA sequence that can change its position within a genome, sometimes creating or reversing mutations and altering the cell's genetic identity and genome size.
  • Transposition often results in duplication of the same genetic material.
  • transposons There are at least two classes of transposons: Class I transposons or retrotransposons generally function via reverse transcription, while Class II transposons or DNA transposons encode the protein transposase, which they require for insertion and excision of DNA
  • nucleic acids may be modified by methyl transferase enzymes, a class of enzymes which can methylate two of DNA's four bases, cytosine and adenine.
  • Adenine or cytosine methylation is part of the restriction modification system of many bacteria. Cytosine methylation is widespread in both eukaryotes and prokaryotes, even though the rate of cytosine DNA methylation can differ greatly between species.
  • nucleic acids may be modified by helicases, which are a class of enzymes that move along a nucleic acid phosphodiester backbone, separating two annealed nucleic acid strands such as DNA and RNA.
  • helicases are a class of enzymes that move along a nucleic acid phosphodiester backbone, separating two annealed nucleic acid strands such as DNA and RNA.
  • Many cellular processes, such as DNA replication, transcription, translation, recombination, DNA repair, and ribosome biogenesis involve the separation of nucleic acid strands that necessitates the use of helicases.
  • Helicases may create nucleic acid fragments.
  • RNA virus detection test kits use a buffer to immediately inactivate at least ambient ribonuclease (RNase) enzyme activity in the biological sample.
  • RNase ambient ribonuclease
  • Low sensitivity test kits for nucleic acids have the problem of false negative results because low levels of nucleic acids in a test sample occur when the test kit procedure uses a buffer which excessively dilutes a test sample and then uses only a fraction of the diluted test sample to detect nucleic acids.
  • One way to inactivate enzymes which destroy RNA and DNA is to use a process of enzyme protein denaturation, commonly by using 5-10 molar urea or a guanidine salt. Urea in high concentrations disrupts the noncovalent bonds in the proteins. Guanidine hydrochloride (also known as guanidinium chloride) has chaotropic properties which denature proteins.
  • a chaotropic agent is a substance which disrupts the structure of, and denatures, macromolecules such as proteins and nucleic acids (e.g. DNA and RNA). Chaotropic solutes increase the entropy of the system by interfering with intermolecular interactions mediated by non-covalent forces such as hydrogen bonds, van der Waals forces, and hydrophobic effects. In aqueous solutions containing 6M guanidine chloride, almost all proteins lose their entire secondary structure and become randomly coiled peptide chains. Guanidine thiocyanate is also used for its denaturing effect on various biological samples (Guanidine, Wikipedia, 2020).
  • virus detection test kits based on detecting nucleic acids include a reducing agent such as about a 0.1 molar concentration of 2-mercaptoethanol (also known as ⁇ -mercaptoethanol, BME, 2BME, 2-ME, ⁇ -met, and HOCH 2 CH 2 SH) to reduce disulfide bonds to irreversibly denature the ribonuclease enzymes in biological samples being tested for the presence of a virus.
  • a reducing agent such as2-Mercaptoethanol is used in RNA isolation procedures to eliminate ribonuclease released during cell lysis by adding it to the sample storage buffer to guarantee the denaturation of the ribonuclease.
  • compositions for detecting viral RNA in a biological sample that does not require unnecessary dilution of the biological sample, does not include unstable and undesirable reducing agents, increased sensitivity, and lower risk of infectivity to healthcare workers and laboratory technicians handling and testing the biological samples.
  • SUMMARY OF THE INVENTION Provided herein are compositions for immediately and efficiently creating surprisingly stable biological samples not needing a reducing agent or refrigeration. The compositions are useful for detecting the presence of viral ribonucleic acid (RNA) in a biological samples.
  • RNA viral ribonucleic acid
  • compositions herein include a lysis buffer having a protein denaturing agent wherein the protein denaturing agent inactivates proteins in the mixture, prevents degradation of nucleic acids, sterilizes the biological sample without the need for refrigeration and wherein the lysis buffer does not include a reducing agent.
  • the compositions protect nucleic acids initially present in the biological sample as a part of the specimen collection process by causing an inhibition of the degradation of the nucleic acids until the biological sample mixture can be tested for a pathogen such as SARS-CoV-2 virus. This inhibition is achieved by including a protein denaturing agent in a lysis buffer of the composition
  • the compositions can be immediately prepared anywhere rapidly and efficiently and safely as the biological matter is being collected.
  • RNA viral ribonucleic acid
  • a lysis buffer including a protein denaturing agent to form a mixture wherein the protein denaturing agent inactivates proteins in the mixture, prevents degradation of nucleic acids, and sterilizes the biological sample
  • the lysis buffer does not include a reducing agent and the method does not require unneeded dilution of the nucleic acids into a fluid, such as VTM, UTM or PBS before contacting the biological sample with a composition of the present invention.
  • compositions and methods of the present invention lessen viral infection risk to a virus sample handler or tester:
  • the lysis buffer is needed to reduce rate of binding or affinity of a coronavirus to a bind and infect a mammalian cell. This would help to make the lysis buffer safer for the person taking the viral sample from that person if contacted then becoming infected with the virus on their body anywhere. This is done by selecting a pH which is not conducive to coronavirus attachment to a cell or replication in a mammalian cell should one remain intact.
  • the denaturing agent in the lysis buffer of the compositions described herein inactivates the virus by denaturing the virus proteins.
  • the compositions of the present invention stabilize the virus RNA from enzymes such as a ribonuclease (RNase).
  • RNase ribonuclease
  • the compositions include one or more cations or anions that bind to the viral RNA and inhibit its degradation by an RNase.
  • hydrolysis of viral RNA is reduced or minimized by controlling the pH of the composition.
  • the compositions of the present invention avoid the use of a reducing agent. Surprisingly, the elimination of reducing agents in the compositions and methods of the present invention does not affect the accuracy of the viral test results.
  • the lysis buffer of the compositions herein are fully effective in lysing cells, denaturing proteins and protecting nucleic acids from ribonucleases, and avoid undesirable characteristics of the reducing agents which are odorous, volatile, unstable (must be bought fresh frequently) and potentially toxic to human use.
  • the concentration of guanidine salt in the lysis buffer of the compositions is minimized to increase the stability of the composition.
  • the lysis buffer has a functional component such as, for example, sodium sulfate, potassium sulfate, sodium citrate, Tris, and/or NaOAc) to provide better COVID RNase and human RNase protein insolubility and minimize degradation of viral RNA in a biological sample.
  • a functional component such as, for example, sodium sulfate, potassium sulfate, sodium citrate, Tris, and/or NaOAc
  • FIGS.1A-1C show a method of biological sample preparation using a flat surface to which is first added biological matter to be lysed by spraying a lysis buffer on the biological matter to form biological sample mixture.
  • FIGS.2A-2C show a method of biological sample mixture preparation using a collection tube in which a lysis buffer is first added to the collection tube and then a biological matter is added to the collection tube to be lysed and to form the biological sample mixture.
  • FIGS.3A-3C depict a method of biological sample mixture preparation using a flat surface to which is first added a lysis buffer, and then added is biological matter to be lysed so as to form a biological sample mixture
  • FIGS.4A-4C depict a procedure wherein a wet nucleic acids sample is added to a porous surface which may be a filter surface of any kind capable of retaining nucleic aci
  • FIG.5A depicts a procedure wherein a collection tube has contents which are a biological sample comprising a lysis buffer fluid with denatured proteins, lysed cell proteins, lipids, and nucleic acids, and optionally further comprising an added nucleic acid.
  • the contents of the sample collection tube 501 comprise a lysis buffer fluid with sample specimen 502, comprising denatured proteins 510 and cellular debris, lysed proteins and lipids 510, and with nucleic acids to be tested 503, and optionally further comprising added nucleic acids 505.
  • FIG.5B depicts performing a non-magnetic isolation of the nucleic acid to be tested 503 by running a spin column 507 having a spin column material 509 in a collection tube 508.
  • Added nucleic acids 505 may be retained along with nucleic acids to be tested 503 in the spin column material 509 or washed and discarded into the flow-through 506 materials which include denatured proteins 510 and cellular debris, lysed proteins and lipids 510.
  • FIG.5C depicts the nucleic acids to be tested 503 and optional added nucleic acid 505 in nuclease-free water 511.
  • FIG.5D depicts an example spin column procedure for purifying RNA and DNA from collection buffer mixture using ethanol for extracting the RNA and DNA and washing the nucleic acids free of the lysis buffer and denatured proteins and cellular debris.
  • FIGS.6A-6D depict a procedure for a magnetic bead-based purification of nucleic acids RNA and DNA from oral swab collected biological matter for SARS-CoV-2 virus.
  • FIGS.7A-7D depict purification of nucleic acids in a mixture of collected biological matter containing a lysis buffer through use of a centrifugal filter membrane.
  • FIGS.8A-8C depict a reverse transcriptase PCR (RT-PCR) process.
  • FIG.9 depicts in accordance with some embodiments of the present invention, several common kinds of biological matter (sample specimen) collection devices.
  • FIGS.10A-10E depict a sequencing procedure to identify nucleic acid mutations for samples giving a positive nucleic acid test result.
  • FIG.11 depicts an overview of the steps in Next Generation Sequencing workflow.
  • FIG.12 is a schematic of steps in the workflow of a next generation sequencing based assay.
  • FIG.13 depicts some of the steps which may be used for some embodiments of the present invention for collecting cellular biological matter specimen in a collection tube, lysing cells in the cellular biological matter specimen in the collection tube, binding RNA from the collected cellular biological matter specimen to a column with a material which binds RNA, and washing and eluting the RNA from the RNA binding column into a tube for holding the RNA in a fluid.
  • FIG.14 depicts aspects of the TaqMan Kit and Process and SYBR Green 1 Reagents used to detect PCR products
  • FIG.15 is a graph showing validation data of the sensitivity of a RELY lysis buffer of the present invention vs PBS (phosphate buffered saline).
  • FIG.16 is a bar graph with PCR Cycles needed on left axis to begin to detect COV-2 Gene N1.
  • FIG.17 is a bar graph providing a data overview of the qPCR results for the SARS-CoV-2 reference RNA gene ‘N1’.
  • FIG.18 is a bar graph providing a data overview of the qPCR results for the SARS-CoV-2 reference RNA gene ‘M’.
  • FIG.19 is a bar graph providing a data overview of the qPCR results for the human control DNA gene RNase P (‘RP’).
  • FIG.20 is a bar graph with PCR cycles on left axis needed for COV-2 Genes N1, M1, and RP tested using a RELY Buffer formulation No.3.
  • FIG.21 is a bar graph showing a data overview for the SARS-CoV-2 three reference genes ‘N1’, ‘M’ and ‘RP’ as tested in PBS.
  • FIG.22 is a graph of storage stability testing conducted with variable storage temperatures on showing how well RELY Lysis Buffer Compared to PBS Buffer acts to protect COV-2 Zeptometrix gene N1.
  • FIG.23 is a graph of storage stability testing conducted with variable storage temperatures on showing how well RELY Lysis Buffer Compared to PBS Buffer acts to protect COV-2 Zeptometrix gene M1. Three temperatures were tested: RT(room temperature approximately 20 ° C), 35 ° C and 50 ° C. DETAILED DESCRIPTION OF THE INVENTION [0063]
  • General embodiments of the present invention comprise testing methods and test kits including immediate use lysis buffer(s) useful for detecting and making various qualitative and quantitative assessments of the presence of virus, bacterial, fungal, animal, and plant biological matter in a test sample.
  • nucleic acids may be obtained from a nucleic acids source selected from the group consisting of a virus, a bacteria, a fungus, an animal, a human, a plant, a lab sample, an environment, a dwelling, a public place, a food sample, a water sample, an air sample, a soil, a synthetic nucleic acids source, and any combination thereof.
  • a nucleic acids sample from biological matter or lab sample is a particularly important source for testing during the ongoing SARS-CoV-2 virus pandemic.
  • a test sample containing nucleic acids may be safely, rapidly, efficiently, accurately detected with high sensitivity, and the nucleic acids may accurately be characterized as a means for identifying a biological source of the nucleic acids.
  • a test kit for detecting SARS-CoV-2 virus nucleic acids needs to safely address obstacles to proper nucleic acids detection including the problem of ambient nucleic acids enzymes in biological samples.
  • compositions use a biological sample collection fluid comprising a CDC-authorized VTM (Virus Transfer Medium), PBS (phosphate buffered saline), Amies transport medium, sterile saline, and a combination thereof during initial biological matter collection, however this is unnecessary for practicing the many embodiments of the present invention.
  • a biological sample collection fluid comprising a CDC-authorized VTM (Virus Transfer Medium), PBS (phosphate buffered saline), Amies transport medium, sterile saline, and a combination thereof during initial biological matter collection, however this is unnecessary for practicing the many embodiments of the present invention.
  • Using the currently existing biological sample collection fluid is responsible for reported SARS-CoV-2 virus test false negative results for several reasons.
  • Collections of, for example, an upper respiratory specimen such as, for example, a nasopharyngeal (NP) specimen, an oropharyngeal (OP) specimen, a nasal mid-turbinate swab, an anterior nares or nasopharyngeal wash/aspirate, or nasal wash/aspirate (NW) specimen from a person who may have become infected with a SARS-CoV-2 virus is a needless dilution of the nucleic acids, the source of which may include nucleic acids from a nucleic acids source selected from the group consisting of a virus, a bacteria, a fungus, an animal, a human, a plant, a lab sample, an environment, a dwelling, a public place, a food sample, a water sample, an air sample, a soil sample, a synthetic nucleic acids source, and any combination thereof.
  • a nucleic acids source selected from the group consisting of a virus, a bacteria,
  • collections of samples which may be a source of nucleic acids selected from the group consisting of a virus, a bacteria, a fungus, an animal, a human, a plant, a lab sample, an environment, a dwelling, a public place, a food sample, a water sample, an air sample, a soil sample, a synthetic nucleic acids source, and any combination thereof.
  • This dilution step is a significant problem that can cause false negative test results in methods attempting to detect the nucleic acids.
  • a swab to collect an upper respiratory specimen such as, for example, a nasopharyngeal (NP) specimen, an oropharyngeal (OP) specimen, a nasal mid- turbinate swab, an anterior nares or nasopharyngeal wash/aspirate or nasal wash/aspirate (NW) specimen from a person who may have become infected with a SARS-CoV-2 virus.
  • NP nasopharyngeal
  • OP oropharyngeal
  • NW nasal wash/aspirate
  • the CDC recommends that the upper respiratory swab specimen should be placed immediately into a sterile transport tube containing 2-3mL of either viral transport medium (VTM), Amies transport medium, or sterile saline. Testing lower respiratory tract specimens is also an option. For patients who develop a productive cough, sputum should be collected and tested for SARS-CoV-2. The induction of sputum is not recommended. When under certain clinical circumstances (e.g., those receiving invasive mechanical ventilation), a lower respiratory tract aspirate or broncho-alveolar lavage sample should be collected and tested as a lower respiratory tract specimen.
  • VTM viral transport medium
  • Amies transport medium Amies transport medium
  • sterile saline sterile saline
  • a non-magnetic process embodiment of the present invention can be used to test for the presence of the virus in an oral swab biological sample taken from a person suspected of being infected with the virus.
  • the sample can be taken from the nose, the mouth, the eyes, the throat, a cut, a wound, from sputum, vomit, excrement, urine, blood, semen, sweat, tears, lymph fluid, or other bodily fluids.
  • the swab can collect a sample from under the tongue, the throat, an oral location, or another region in the body which may be infected or contaminated with the virus.
  • a sample does not necessarily have to originate from a swab, it can also be collected from saliva, spit, sputum, a syringe needle biopsy or from a sample obtained from the body of a deceased person.
  • the reagent composition of the present invention or the lysis buffer may be applied onto a wall or another surface of a support material (such as paper) or container (such as a collection tube) in liquid or dry form, or be applied for instance as a spray.
  • the present invention method embodiments and compositions may comprise using a lyophilized lysis buffer composition that stabilizes the biological sample upon contact, such as within 15-30 seconds or more generally 5-60 seconds after allowing a person to spit into a dry collection tube that comprises the lysis buffer.
  • a lyophilized lysis buffer composition that stabilizes the biological sample upon contact, such as within 15-30 seconds or more generally 5-60 seconds after allowing a person to spit into a dry collection tube that comprises the lysis buffer.
  • the invention embodiments provide various compositions comprising a lysis buffer and may further comprise a salt.
  • the lysis buffer may be immobilized on a tissue, paper, tube, or swab.
  • a person may cough or spit on invention embodiments which provide the various compositions of the invention which comprise a lysis buffer and may further comprise a salt, wherein the lysis buffer and or lysis salts are present as a solid, a gel, a damp area, a fluid, or in a releasable formulation immobilized or loosely present on or wetting for example a tissue, a paper, a tube or a swab which function as a method using a kit comprising a sampling device or sample receptacle providing a lysis means for constituents comprising the biological matter which may contain an amount of an infectious pathogen such as a virus such as SARS-CoV-2.
  • Such a receptacle for a biological specimen or sample due to the presence of a lysis buffer embodiment of the present invention can easily be shipped without having to refrigerate the receptacle for a biological specimen or sample.
  • lysis medium or even dry lysis composition from a foil or plastic container could be added immediately to the sample container or collection tube, receptacle, or biological sample soon after a person provided the specimen of biological matter or the lysis composition may be delivered as a spray onto the biological specimen which may contain the infectious pathogen such as SARS-CoV-2.
  • the practicing methods and compositions of the lysis buffer may further comprise an added nucleic acid optionally comprising a unique genetic sequence that are included in the biological sample or lysis buffer composition used.
  • An added nucleic acid is a term for a chemical marker which can be added and used as a method embodiment of the present invention comprising: an internal control for verifying correct, accurate and safe operation of the sample collection and testing procedure, tracking the biological sample during biological sample preparation; shipping or transporting the biological sample; testing the biological sample; reporting results of the biological sample; and organizing results of various tests of the biological sample by same or multiple labs, and facilitating tested persons’ HIPAA (Health Insurance Portability and Accountability Act of 1996) rights to anonymous protection of the tested persons health test data, and safe dissemination of the test data in a publication or for demographics regarding the COVID-19 pandemic.
  • HIPAA Health Insurance Portability and Accountability Act of 1996) rights to anonymous protection of the tested persons health test data, and safe dissemination of the test data in a publication or for demographics regarding the COVID-19 pandemic
  • lysis buffer embodiments of the present invention can be prepared and stored over weeks, months, or years before it is being used.
  • Virus test kits use biological samples which detect virus nucleic acids, RNA (ribonucleic acid), or DNA (deoxyribonucleic acid) depending on the target virus or microorganism.
  • RNA and DNA are polymers composed of nucleotide monomers which each have three components: a 5-carbon sugar, a phosphate group, and a nitrogenous base.
  • the sugar in an RNA nucleotide is ribose and is deoxyribose in a DNA nucleotide.
  • Strings of nucleotides are bonded to form helical backbones - typically, one for RNA, two for DNA - and assembled into chains of base-pairs selected from the five primary, or canonical, nucleobases: adenine, cytosine, guanine, thymine, and uracil. Thymine occurs only in DNA and uracil only in RNA.
  • the sugars and phosphates in nucleic acids are connected to each other in an alternating chain (sugar-phosphate backbone) through phosphodiester linkages.
  • the carbons to which the phosphate groups attach are the 3'-end and the 5'-end carbons of the sugar. Ends of nucleic acid molecules are called the 5'-end and the 3'-end.
  • the nitrogenous bases (nucleobases) are bonded to the sugars via an N-glycosidic linkage between a nucleobase ring nitrogen (N-1 for pyrimidines and N-9 for purines) and the 1' carbon of the sugar.
  • RNA virus and DNA virus detection test kits rely upon being able to identify a portion of the RNA sequence of an RNA virus or a portion of the DNA sequence of a DNA virus.
  • RNA and DNA biopolymers found in animals, bacteria, archaea, mitochondria, chloroplasts, and viruses range in size from 21 nucleotides, such as small interfering RNA (siRNA), to large chromosomes in a single molecule that contain 247 million base pairs (human chromosome 1). Most living cells contain both DNA and RNA, with the exception of cells such as mature red blood cells. Viruses usually contain either DNA or RNA, but not both. If the virus is an RNA virus, then a reverse transcriptase enzyme process must be performed to prepare the corresponding DNA reverse transcript. After this, the DNA in the biological sample can be amplified (mass-produced) using a PCR (polymerase chain reaction) process.
  • PCR polymerase chain reaction
  • Some embodiments of the invention comprise an improved testing method for performing a DNA virus nucleic acid detection test on a biological sample, the improved testing method comprising the steps of: (a) obtaining a biological sample from a selected source of biological matter; (b) contacting the biological sample without delay with an amount of lysis buffer to form a mixture, wherein the lysis buffer comprises a protein denaturing agent in a fluid; (c) using the protein denaturing agent from the fluid lysis buffer in the mixture as a means to denature proteins immediately without refrigeration and keep the proteins inactivated in the mixture and prevented from degrading nucleic acids in the mixture; (d) using the protein denaturing agent from the fluid lysis buffer in the mixture to cause an immediate lysis of biological cells, micro
  • Example 2 An embodiment of the present invention methods and compositions for testing for RNA virus in a person
  • Some embodiments of the invention comprise an improved testing method for performing an RNA virus nucleic acid detection test on a biological sample, the improved testing method comprising the steps of: (a) obtaining a biological sample from a selected source of biological matter; (b) contacting the biological sample without delay with an amount of lysis buffer to form a mixture, wherein the lysis buffer comprises a protein denaturing agent in a fluid; (c) using the protein denaturing agent from the fluid lysis buffer in the mixture as a means without refrigeration to denature proteins immediately and keep the proteins inactivated in the mixture and prevented from degrading nucleic acids in the mixture; (d) using the protein denaturing agent from the fluid lysis buffer in the mixture to cause an immediate lysis of biological cells, microorganisms, bacteria, virus particles, and any other infectious organisms that may have come from the biological matter in the biological sample so as to sterilize the mixture immediately and safely; (e)
  • Example 3 Embodiment of the present invention test methods and compositions for testing for nonhuman DNA in a person
  • Some embodiments of the invention comprise an improved testing method for performing a non-human DNA detection test on a biological sample, the improved testing method comprising the steps of: (a) obtaining a biological sample from a selected source of biological matter; (b) contacting the biological sample without delay with an amount of a lysis buffer to form a mixture, wherein the lysis buffer comprises a protein denaturing agent in a fluid; (c) using the protein denaturing agent from the fluid lysis buffer in the mixture as a means without refrigeration to denature proteins immediately and keep the proteins inactivated in the mixture and prevented from degrading nucleic acids in the mixture; (d) using the protein denaturing agent from the fluid lysis buffer in the mixture to cause an immediate lysis of biological cells, microorganisms, bacteria, virus particles, and any other infectious organisms that may have come from the biological matter in the biological sample so as to sterilize the mixture immediately and safely;
  • Example 4 Another embodiment of the present invention provides test methods and compositions for testing for nonhuman RNA in a person.
  • Some embodiments of the invention comprise an improved testing method for performing a nonhuman RNA detection test on a biological sample, the improved testing method comprising the steps of: (a) obtaining a biological sample from a selected source of biological matter; (b) contacting the biological sample without delay with an amount of a lysis buffer to form a mixture, wherein the lysis buffer comprises a protein denaturing agent in a fluid; (c) using the protein denaturing agent from the fluid lysis buffer in the mixture as a means without refrigeration to denature proteins immediately and keep the proteins inactivated in the mixture and prevented from degrading nucleic acids in the mixture; (d) using the protein denaturing agent from the fluid lysis buffer in the mixture also to cause an immediate lysis of biological cells, microorganisms, bacteria, virus particles, and any other infectious organisms that may have come from the biological matter in the biological sample so as to sterilize the mixture immediately and safely; (e)
  • Some embodiments of the present invention are designed to provide high- sensitivity true positive results, and to not provide false negative results.
  • Some embodiments of the invention comprise buffer compositions useful as a safe viral lysis sample collection and testing buffer. After safely lysing the nucleic acids, then the identity of the nucleic acids can be tested using various nucleic acid tests using either known or novel chemical, biochemical and genetic biotechnology procedures which may include a selected PCR procedure, a colorimetric process, and/or a nucleic acid sequencing process with reference nucleic acids sequence(s) of a known genus, species, or mutant species of a life form or a virus.
  • FIG.1 depicts some initial steps of one embodiment of the present invention which is one method of adding a lysis buffer 110 as a spray 108 or as a buffer mist 108 onto a dry nucleic acids sample 109 on a surface 107 as a means for protecting nucleic acids to be tested 103 from various enzymes of nucleic acids (see Background for examples of the enzymes) as well as lysing viral capsules, bacterial walls, cell membranes or other membranes which may be confining the nucleic acids to be tested 103 in the sample 102, 106, or 109.
  • a lysis buffer 110 as a spray 108 or as a buffer mist 108 onto a dry nucleic acids sample 109 on a surface 107 as a means for protecting nucleic acids to be tested 103 from various enzymes of nucleic acids (see Background for examples of the enzymes) as well as lysing viral capsules, bacterial walls, cell membranes or other membranes which may be confining the nucleic acids to be tested
  • one method of delivering a spray formulation 108 of the lysis buffer 110 as a means for protecting nucleic acids to be tested 103 from various enzymes of nucleic acids may comprise the steps of: (a) depositing a wet sample specimen 102 with nucleic acids to be tested 103 upon a surface 101; (b) allowing optionally, the wet sample droplet 102 to situate as a wet sample 106 of the nucleic acids to be tested 103 on a surface 104 for a period of drying (wherein surface 104 which may be surface 101); and then (c) adding a lysis buffer 110 which comprises a formulation 108 by spraying from a container nozzle tip 111 upon a dried, semi-dry, or wet nucleic acids sample 109 which is situated on a surface 107 (which may have been the same surface as surface 101, or surface 104).
  • the source of the wet nucleic acids sample specimen 102 may be a biological sample from any location or body biological material of a person or an animal.
  • one or more added nucleic acid in the physical form of an oligonucleotide 105 may be added to the original nucleic acids sample to serve as an internal control, for relative quantification of copy numbers between the nucleic acid to be tested and the added nucleic acid(s), for quality control and tracking, for calibration of false negative or false positive sensitivity and probability, as a carrier nucleic acid, carrier RNA, carrier DNA, and any combination thereof, or for other purposes.
  • a biological sample which contains nucleic acids to be tested 103 may come from a living or dead human, animal, or be an in vitro biological sample, or might be from an inanimate location.
  • the location of the biological sample containing nucleic acids to be tested 103 may be selected from the group consisting of a skin location, a hair, a wound, a sore, a blister, a scab, a finger nails scraping sample, a sweat sample, an eye, an inside cheek surface, a tongue surface, inside a nostril, throat, from another oral cavity location, a urine sample, a fecal matter sample, a blood sample, a blood plasma sample, a lymph sample, a cerebrospinal fluid sample, a sputum sample, a saliva sample, a vomit sample, a respiratory tract fluid or surfactant liquid sample, a human sample, an animal sample, a bone marrow sample, an internal organ, a body secretion,
  • FIG.1 is an example procedure wherein method of biological sample mixture preparation uses a flat surface to which first added is biological matter to be lysed by spraying a lysis buffer on the biological matter to form biological sample mixture.
  • FIG.1A depicts an example procedure wherein a wet sample specimen 102 comprising a nucleic acid to be tested 103 is provided, adding the wet sample specimen 102 comprising a nucleic acid to be tested 103 to a surface 101 and allowing it to dry or remain wet on a paper surface 104 as shown in FIG.1B, and in step (C) then spraying the components depicted in FIG.1B with a lysis buffer mist 108.
  • the lysis buffer 108 may further comprise an added nucleic acid 105.
  • the added nucleic acid 105 may comprise an internal control, a carrier nucleic acid, carrier RNA, carrier DNA, and any combination thereof.
  • a biological sample which contains nucleic acid to be tested 103 may come from a living or dead human, animal, or be an in vitro biological sample, or might be from an inanimate location.
  • the location of the biological sample containing nucleic acid to be tested 103 may be selected from the group consisting of a skin location, a hair, a wound, a sore, a blister, a scab, a finger nails scraping sample, a sweat sample, an eye, an inside cheek surface, a tongue surface, inside a nostril, throat, from another oral cavity location, a urine sample, a fecal matter sample, a blood sample, a blood plasma sample, a lymph sample, a cerebrospinal fluid sample, a sputum sample, a saliva sample, a vomit sample, a respiratory tract fluid or surfactant liquid sample, a human sample, an animal sample, a bone marrow, an internal organ a body secretion, a tissue biopsy, a cell sample, a corpse, and any combination thereof.
  • the protein concentration in saliva samples is 0.5-1.5 mg/ml. Assuming there is less than 1 ml saliva on the collection swab, then the protein amount is below 1 mg, and therefore the protein concentration in the lysis buffer can be below 1 mg protein/ml biological sample mixture (see FIG.2 description).
  • the surface 101, surface 104, and surface 107 may be the same or different surfaces, and may be selected from the group consisting of a dry surface, a solvent-wetted surface, a surface wetted lysis buffer 110, a surface wetted with a large aqueous concentration of known-sequence polynucleotides as a polynucleotides or nucleic acid chum, a paper surface, a wax paper surface, a plastic surface, a test tube surface, a filter membrane surface, an Amicon filter surface, an Amicon filter surface for retaining polynucleotides with a length of greater than 100 nucleic acids bases on the surface of the Amicon filter surface, a 3K Amicon filter surface, a 10K Amicon filter surface, a 50K Amicon filter surface, a Millipore filter surface, an Millipore filter surface for retaining polynucleotides with greater than
  • the term ‘chum’ refers to carrier nucleic acids, such as carrier RNA or carrier DNA, which are added to the sample or reaction vessel to stabilize the actual biological sample or nucleic acid of interest.
  • carrier RNA is useful to reduce or prevent degradation of the RNA that is to be tested for.
  • residual enzymes acting on nucleic acids, such as RNases which may be present in the collected biological sample (or other mechanisms that result in the general degradation, adsorption or inactivation of nucleic acids, DNA or RNA) will be engaged by the chum.
  • Chum is advantageously added at high concentrations for the purpose of protecting the nucleic acid, DNA or RNA that is to be tested for from degradation, adsorption or inactivation.
  • concentrations of chum such as added carrier RNA
  • concentrations of chum are 10 pM – 1 nM, 1 nM – 100 nM, 100 nM – 10 ⁇ M, 10 ⁇ M – 1 mM and any combination thereof.
  • the added chum or carrier RNA can alternatively be provided in the reagent buffer or in any other reaction component that is part of the testing procedure, such as in a tube or vessel that is pre-coated or pre- equipped with carrier RNA, positive or negative controls, or other blocking agents that stabilize RNA, DNA or nucleic acids from degradation, adsorption or inactivation.
  • nucleic acid to be tested 103 containing samples may be handled as depicted in FIG.1 and air-dried on a surface 101, a surface 104, or a surface 107 before or after spraying the immobilized sample specimen 106 with the lysis buffer mist 108.
  • the nucleic acid to be tested 103 sample may be dried by using a drying step comprising spray-drying, freeze-drying, using a desiccant or drying in a vacuum bell jar under vacuum.
  • a refrigerating step may be useful for preserving some nucleic acids samples, the refrigerating step comprising storing the nucleic acid samples in a conventional refrigerator/freezer, storing them on ice or dry ice in a cooler, or storing in a subzero refrigerator or storing in a container cooled by liquid nitrogen.
  • FIG.2 depicts another example procedure embodiment of the present invention wherein the method of biological sample mixture preparation utilizes a collection tube in which first added is lysis buffer and then added is biological matter to be lysed to form the biological sample mixture.
  • a lysis buffer 208 optionally containing an added nucleic acid 205 is added to a biological sample collection tube 201.
  • a nucleic acid to be tested 203 in biological matter such as fluid of a sample specimen 202 is added to collection tube 201 for immediately creating a biological sample mixture depicted in step (C) of FIG.2 which comprises the lysis buffer 208 and sample specimen 202, optionally further comprising an added nucleic acid 205, all of which are added to the sample collection tube 201.
  • an added nucleic acid may comprise an internal control, a quality control, a negative control, a carrier nucleic acid, carrier RNA, carrier DNA, and any combination thereof.
  • the biological sample mixture depicted in step (C) preferably is created essentially at the same time as the sample specimen 202 is collected from a person, using for example a collection swab.
  • Present invention embodiments of the biological sample mixture may be of a great advantage in settings and countries that do not have refrigeration.
  • Collection tubes 201 in steps (A), (B), and (C) preferably are the same collection tube 201.
  • This fluid mixture of the lysis buffer 208 and specimen 202, and optionally with an added nucleic acid 205, is a novel composition embodiment of the present invention and a method comprising: creating immediately and efficiently a surprisingly stable biological sample not needing refrigeration.
  • This fluid mixture is a nonobvious composition and method preparation step in view of the worldwide practices taught in 2020 by all health care emergency doctors, labs, CDC, and WHO officials and doctors whose edicts and standard operating procedures (SOP) for biological sample preparation for dealing with SARS-CoV-2 testing are quite different.
  • inventor(s) have invented a biological sample mixture which usefully does not need refrigeration (refrigeration free method) as a means for effectively protecting any nucleic acids initially present in the biological sample by causing an inhibition of the degradation of the nucleic acids until the biological sample mixture can be tested for a pathogen such as SARS-CoV-2 virus.
  • FIG. 2 depicts a method for forming a composition which is a mixture which causes an inhibition of both possible rapid degradation and possible slow degradation of the nucleic acids in the biological sample from a biological matter of a person. This inhibition is caused by including a protein denaturing agent in the lysis buffer which acts immediately in the fluid mixture depicted in rightmost collection tube 201 depicted in FIG.2.
  • Such embodiments of the present invention are nonobvious and novel biological sample mixture compositions and sample preparation methods because the currently available testing procedures do not use a lysis buffer and require refrigeration before and during shipping, transporting, and storing the prior art biological samples prepared until there is a time to perform a pathogen detection test using a PCR method which in the prior art depends upon PCR equipment.
  • currently available methods involve unneeded dilution of the nucleic acids concentration in the collected biological specimen by an estimated between 1.25- fold to 5-fold. This causes prior art PCR tests more likely to report a false negative test result. Over dilution of the biological sample lowers test sensitivity and accuracy particularly if the person’s biological specimen is problematic in having low virus titer to start with.
  • FIG.2 depicts a method for forming a composition which is a mixture which causes an inhibition of both possible rapid degradation and possible slow degradation of the nucleic acids in the biological sample from a biological matter of a person.
  • This inhibition is caused by the lysis buffer 208 which comprises a fluid with a protein denaturing agent.
  • the sample specimen mixture is a fluid in a collection tube 201 which would be in rightmost collection tube 201 depicted in FIG.2.
  • the lysis buffer 208 is capable of lysing cell walls of cells in the biological matter of sample specimen 202.
  • FIG.3 shows an example procedure wherein the method of biological sample mixture preparation uses a flat surface to which first added is a lysis buffer and then added is biological matter to be lysed so as to form a biological sample mixture
  • FIGS.3A-3C Depicted in FIGS.3A-3C is an example procedure wherein method of biological sample mixture preparation is a step of using a flat surface which is the same surface numbered 301, 304 and 307.
  • FIG.3 step (A) is a step for adding the lysis buffer 308 optionally containing an added nucleic acid 305 to a flat surface 301 which is capable of absorbing or drying the lysis buffer 308.
  • step (B) is a step for adding a sample specimen 302 containing some nucleic acids to be tested 303 to surface 304 which already contains an amount of lysis buffer 308 which is labeled in step (B) as lysis buffer 309, applied or immobilized to flat surface 301.
  • step (C) depicted is the biological sample mixture composition which will have immediately denatured proteins and enzymes in the biological sample mixture as well as have lysed cell walls and organelle walls inside cells so as to permit a complete inactivation of ribonucleases, deoxyribonucleases, and other enzymes which if not inactivated by denaturing them, could rapidly degrade the nucleic acids from sample specimen 302.
  • the lysis buffer 308 is applied to personal protective equipment (PPE), such as surgical masks, air filtration devices or air filters, shields, suits, gloves, shoes, gowns, either worn by a person as part of PPE or installed in instruments or facilities, such that SARS-CoV-2 containing aerosols or particles are inactivated upon contact with the PPE.
  • PPE personal protective equipment
  • Protective equipment as defined here includes PPE as well as systems that protect more than one individual. Examples include systems for heating, ventilation, air conditioning / HVAC, air filtration, sterilization, recirculation of breathable air for use either inside or outside.
  • the lysis buffer may comprise reagents that undergo a visible color change after a swab, sputum or another biological sample containing the virus or the nucleic acid to be tested for is detected. Such a color change is useful to indicate a "SAFE" time after which the biological sample is inactivated and no longer infectious after being brought in contact with the lysis buffer or the dried/lyophilized components of the lysis buffer.
  • the color change reaction may occur in the collection tube after the biological samples is added, on PPE, such as on a protective mask worn by a person, when she comes in contact with the biological sample, the virus or the nucleic acid to be tested for, or on equipment or instrumentation that may come in contact with the biological sample, the virus or the nucleic acid to be tested.
  • FIG.4 Procedure wherein wet nucleic acids sample is added to a porous surface which may be a filter surface of any kind capable of retaining nucleic acids
  • step (A) depicts an example procedure step for adding nucleic acid to be tested 403 sample specimen 402 to a porous surface 401 which may be a filter surface of any kind capable of retaining nucleic acids to some extent.
  • a sample specimen 402 optionally is allowed to dry or is allowed remain wet on a filter surface 404as shown in step (B) and then in step (C) added is a lysis buffer 408 optionally containing an added nucleic acid405 with the immobilized biological sample mixture 406_depicted in step (C).
  • Filter surfaces 401, 404, and 407 may be same or different or/and may be flat or curved in any conceivable manner.
  • the lysis buffer 408 is for denaturing proteins in the biological sample mixture 40_ so as to inactivate ribonucleases, deoxyribonucleases and other related nucleic acid degrading enzymes likely present in the wet nucleic acids to be tested 403.
  • FIG.5A Procedure wherein a collection tube has contents which are a biological sample comprising a lysis buffer fluid with denatured proteins, lysed cell proteins, lipids, and nucleic acids, and optionally further comprising an added nucleic acid.
  • FIG.5A depicts an example procedure step for placing into a sample collection tube 501 contents which comprise a lysis buffer fluid with sample specimen 502, comprising denatured proteins 510 and cellular debris, lysed proteins and lipids 510, and with nucleic acids to be tested 503, and optionally further comprising added nucleic acids 505.
  • FIG.5 B depicts performing a non-magnetic isolation of the nucleic acid to be tested 503 by running a spin column 507 having a spin column material 509 in a collection tube 508.
  • FIG.5C depicts the nucleic acids to be tested 503 and optional added nucleic acid 505 in nuclease-free water 511.
  • FIG.5D depicts an example spin column procedure for purifying RNA and DNA from collection buffer mixture using ethanol for extracting the RNA and DNA and washing the nucleic acids free of the lysis buffer and denatured proteins and cellular debris.
  • Another spin column procedure for purifying nucleic acids comprises the steps of: 1) adding a volume of absolute ethanol (100%) equivalent to 0.8x to 2x of the volume of the biological sample mixture (comprising the biological sample and lysis buffer and optionally an added nucleic acid) to the sample collection tube and mixing the sample of nucleic acids in the collection tube by vortexing; [00109] 2) transferring the mixture into a spin column in a collection tube 508 and centrifuging at >10,000 x g for one minute, then discarding the collection tube including the flow-through fluid; [00110] 3) transferring the spin column containing the spun down sample to a new collection tube and adding a volume between 400-700 ⁇ l ethanol (80%) to the spin column and centrifuging at 10,000 x g for one minute, then discarding the flow- through fluid; [00111] 4) adding a volume between 400-700 ⁇ l ethanol (80%) to the spin column and centrifuging at >10,000 x g for one minute, then discard
  • the spin column procedure for purifying nucleic acids instead of using ethanol, may use a hydroxylated organic chemical selected from the group consisting of isopropanol, methanol, n-propanol, a butyl alcohol, a pentyl alcohol, a hexyl alcohol, ethylene glycol, propylene glycol, glycerol, and any combination thereof.
  • a hydroxylated organic chemical selected from the group consisting of isopropanol, methanol, n-propanol, a butyl alcohol, a pentyl alcohol, a hexyl alcohol, ethylene glycol, propylene glycol, glycerol, and any combination thereof.
  • an alternative type of alcohol, polyalcohol or dehydrating agent can be used, such as toluene, nitromethane, ether and any combination thereof.
  • FIG.6 Procedure for a magnetic bead-based purification of nucleic acids RNA and DNA from oral swab collected biological matter for SARS-CoV-2 virus
  • FIG.6 depicts an example of a procedure for using a magnetic bead-based methods for purifying nucleic acids (RNA and DNA) from oral swabs collection of a biological matter for testing for SARS-CoV-2 virus particles (virions).
  • RNA and DNA nucleic acids
  • viral particles virus particles
  • One example of magnetic particles for the separation and purification of nucleic acids are described in: Magnetic particles for the separation and purification of nucleic acids, Berensmeier S, Applied Microbiology and Biotechnology volume 73, pages 495–504 (2006).
  • step (C) but not depicted is a present invention example embodiment method step comprising first creating a biological sample by situating some biological material (not depicted) in form of a sample specimen collected by a collection means such as for example using a specimen swab (not depicted) into sample collection tube 601 and then adding an amount 100-1000 ⁇ l of a lysis buffer so that the sample collection tube 601 contains nucleic acids to be tested 603, denatured enzymes such as ribonucleases and deoxyribonucleases 610, cell debris, other denatured proteins 610, and lysed cells 610.
  • the specimen swab (not depicted in FIG.
  • FIG.6A Depicted in FIG.6A is a step of adding reagent 604 containing magnetic beads 612 and optionally the added nucleic acid 605 to the collection tube 601 containing the sample specimen 602, which contains the nucleic acid to be tested 603 as well as cellular debris and other unwanted components 610.
  • the mixture in collection tube 601 is mixed by pipetting it up and down, during which time the magnetic beads 612 bind to the nucleic acid to be tested 603 and optionally to the added nucleic acid 605 or control nucleic acids if present.
  • Depicted in FIG.6B is a transferring of the collection tube 601 onto a magnetic rack 613 where the contents in the collection tube 601 are magnetized for 5 min or until the solution inside collection tube 601 appears clear.
  • the magnetic rack 613 functions to pull the complex 614 comprising the magnetic beads 612, nucleic acid to be tested 603 and optionally the added nucleic acid 605 or control nucleic acids onto the inside wall of collection tube 601.
  • Supernatant from collection tube 601 is removed (arrow 615) and discarded without disturbing the complex 614 comprising the bead pellet with nucleic acid to be tested 603 and optionally the added nucleic acid 605. Then (arrow 616) about 100 – 1000 ⁇ l (microliters) ethanol 617 are added to each collection tube 601. Do not mix. Then the supernatant containing ethanol is removed from collection tube 601 (see arrow 615) and discarded without disturbing the complex 614. Repeat this procedure. Then remove the collection tube 601 from the magnetic rack 613 and air dry the pellet complex 614 for 5-10 min.
  • FIG.6C Depicted in FIG.6C is the step of adding (arrow 619) of a volume of between about 10 ⁇ l to about 100 ⁇ l of a nuclease-free water 618 to each collection tube 601 and then there is a pipetting of the sample up and down until the complex 614 is dissolved. Incubate the collection tube 601 for 5 min. Transfer the collection tube 601 containing complex 614 onto the magnetic rack 613 and incubate the sample for 2-10 min. or until the supernatant solution appears clear.
  • FIG.6D Depicted in FIG.6D is the step of aspirating (see arrow 620) the supernatant 622 comprising the eluted nucleic acids to be tested 603 optionally the added nucleic acid 605 to a new collection tube 621.
  • the eluted nucleic acids to be tested 603 and optionally the added nucleic acid 605 can be used immediately for molecular based applications or stored at -80oC for future use.
  • FIG.7 Purification of nucleic acids in a mixture of collected biological matter containing a lysis buffer through use of a centrifugal filter membrane [00121] Depicted in FIGS.7A-7D are method steps for purifying nucleic acids in a biological collection mixture through use of a 10-100 kDa centrifugal filter membrane. Nucleic acids that are larger than 50-500 base pairs that be retained.
  • FIG.7A Depicted in FIG.7A is adding the sample specimen 702 containing cellular debris and other unwanted components as well as comprising the nucleic acid to be tested 703 and optionally the added nucleic acid 705 (with 703 and 705 together herein being collectively referred to as the nucleic acid batch 704) to a centrifugal filter membrane device 706 and insert it into an empty collection tube 701 with a cap 707.
  • step (B) is spinning the collection tube 701 containing the centrifugal filter membrane device 706 and the nucleic acid batch 704 in order to force the sample specimen 702 through the centrifugal filter membrane 709, thereby separating the sample specimen 702 into a retained fraction 708 comprising the nucleic acid batch 704, which is being retained by the centrifugal filter membrane 709, and the flow-through 710 containing cellular debris and other unwanted components, which passes the centrifugal filter membrane 709 and is collected at the bottom of tube 701. Discard the collection tube 701 and the flow-through 710.
  • FIG.7C Depicted in FIG.7C is process including inverting the centrifugal filter membrane device 706, inserting it into a new empty collection tube 711 and spinning for recovering the retained fraction 708 comprising the nucleic acid batch 704 into collection tube 711.
  • FIG.7D Depicted in FIG.7D is removing the centrifugal filter membrane device 706, for retaining collection tube 711 comprising the retained fraction 708 with the nucleic acid batch 704.
  • FIG.8B depicts a reverse transcriptase PCR (RT-PCR) process using viral RNA: [00129]
  • FIG.8C depicts a reverse transcriptase PCR (RT-PCR) process.
  • FIG.9 below depicts in accordance with some embodiments of the present invention, several common kinds of biological matter (sample specimen) collection devices. Depicted are: a device which is collection swab 901; a device 902 which is a collection swab 904 in a protective test tube 903 for protecting the collection swab 904; a device which is a test tube 907 with cork 906 to hold a fluidic biological matter specimen 908; and a device which is a syringe 909 to draw fluid samples from a person’s body, a liquid biological matter specimen 910, a biopsy sample 912 on a microscope slide or surface 911, or a biopsy sample 913.
  • Biological matter from the collection device is preferably immediately (without delay) added to a lysis buffer to more effectively accomplish various objects of the present invention, including: (1) using the lysis buffer for inactivating any infectious pathogen in the biological matter sample specimen, and (2) using the lysis buffer for lysing cells and denaturing various proteins including enzymes and the like as a means for inhibiting degradation of the nucleic acids which may be present in the biological matter sample specimen.
  • these objects of the present invention are accomplished by forming a mixture which comprises the biological sample with the lysis buffer, then (3) the mixture being lysed by the lysis buffer need not be refrigerated (to inhibit enzymes such as ribonucleases and preserve the biological cells and the nucleic acids in the sample).
  • the nucleic acids in the lysed biological matter sample can be transported, shipped, stored from any collection without the need for dry ice or ice. This for example frees up the collection sampling in remote third world locations, and even would allow self-sampling by a person needing to know if they have a SARS- CoV-2 virus infection.
  • the mixture also represents a useful means for performing a minimal dilution of the nucleic acids in the biological matter specimen step as a means for allowing subsequent steps of running the nucleic acids detection testing method to be performed with a larger amount of the nucleic acids of the biological matter specimen.
  • FIG.10 depicts a sequencing procedure to identify nucleic acid mutations for samples giving a positive nucleic acid test result.
  • the heat stable Taq (‘AmpliTaq’) enzyme has a 5’-exonucleolytic function that does not displace the probe sitting in the middle but instead digests it, releasing the reporter dye in the process.
  • Phi29 is a polymerase that is not heat stable (40C max) but which has a strand displacement function.
  • Phi29 and related polymerases are used for isothermal amplification processes such as whole-genome amplification (WGA) or Loop-mediated isothermal amplification (LAMP).
  • Bst DNA Polymerase is a thermoresistant polymerase primarily used for Loop-Mediated Isothermal Amplification (LAMP) that has been modified to eliminate 5 ⁇ ⁇ 3 ⁇ exonuclease activity while retaining its 5 ⁇ ⁇ 3 ⁇ polymerase activity at elevated temperatures, around 65°C. It is suitable for applications requiring thermophilic strand displacement polymerase activity and gap-filling reactions and provides high product yields in demanding conditions.
  • LAMP Loop-Mediated Isothermal Amplification
  • oligonucleotide primers that have not been protected from 3’-exonuclease digestion, such as by the addition of one or more phosphorothioate bonds during oligonucleotide synthesis, thereby making the synthesis of such oligonucleotides considerably cheaper.
  • a phosphorothioate bond substitutes a sulfur atom for a non-bridging oxygen in the phosphate backbone of an oligonucleotide. This modification renders the inter- nucleotide linkage resistant to nuclease degradation.
  • Phosphorothioate bonds can be introduced between the last 3-5 nucleotides at the 5'- or 3'-end of the oligonucleotide to inhibit exonuclease degradation.
  • FIG.11 depicts an overview of the steps in Next Generation Sequencing workflow.
  • FIG.12 is a schematic of steps in the workflow of a next generation sequencing based assay.
  • Control Test Samples Needed for Nucleic Acids Testing [00139] Various standard controls need to be run by the testing procedure, including testing known amounts of nucleic acids in an internal control test sample.
  • An internal control test is a specific sequence of nucleic acid, typically DNA or RNA, that is added to the testing procedure at a known concentration or copy number to ensure that the testing procedure works reliably and is able to detect both the positive control(s) and the sample at hand with the expected sensitivity.
  • Exogenous internal controls refer to the use of external DNA or RNA carrying a target of interest. If these positive controls are assayed in separate wells/tubes from the experimental sample, they serve as a control to determine whether the reverse transcription and/or PCR reaction conditions are optimal or not. Additionally, exogenous DNA or RNA positive controls may be spiked into the experimental sample(s) and assayed in parallel or in a multiplex format with the target of interest. These control reactions assess whether the samples contain any components that inhibit reverse transcription and/or PCR.
  • an exogenous control in a sample of a patient being tested for SARS-CoV-2 is nucleic acid of a specific sequence that is added or spiked into the reaction in which the testing is performed.
  • an exogenous control can comprise one or more artificial sequences of nucleic acid, DNA or RNA that do not occur naturally in the biological samples being tested. It is advantageous to include several exogenous controls for reliability and redundancy. In order to obtain a more accurate estimate of the patient’s virus titer or copy number, it is further advantageous to include several exogenous controls at different concentrations or copy numbers, thereby allowing for a direct comparison of viral or pathogen load based on the titers or copy numbers of the various exogenous control or controls.
  • an exogenous control is provided at the following concentrations: 1 - 100fM, 100fM - 10pM, 10pM – 1nM, 1nM – 100nM, 100nM – 10 ⁇ M, 10 ⁇ M – 1mM, and any combination thereof.
  • An internal or negative control can be provided either in liquid or dried form. Several internal or negative controls can be provided in combination with either the same or at different concentrations or copy numbers.
  • Endogenous internal controls refer to the use of a native target that is present in the experimental sample(s) of interest but is different from the target under study. These types of controls are often referred to as normalizers, and are typically used to correct for quantity and quality differences between samples.
  • One example of an endogenous control in a sample of a patient being tested for SARS-CoV-2 is nucleic acid that is already present in the patient’s saliva, sputum, bodily fluid, tissue or from any other of the types and sources of biological samples described herein, but not originating from the actual SARS-CoV-2 virus.
  • an endogenous control can be useful to determine and quantify the efficiency of the biological sample collection, in other words as a measure of how effectively endogenous and viral material was acquired during the sample collection procedure. Doing so can provide a more accurate estimate of the patient’s virus titer or copy number through normalization based on the titer or copy number of the endogenous control locus or loci.
  • an endogenous control is selected from a suitable nucleic acid that is present at the following concentrations: 1 - 100fM, 100fM - 10pM, 10pM – 1nM, 1nM – 100nM, 100nM – 10 ⁇ M, 10 ⁇ M – 1mM and any combination thereof.
  • reaction wells may include assays that detect artificial positive or negative controls spiked into each sample during a cDNA synthesis step, thereby ensuring that the reverse transcription step proceeded as needed.
  • Virus Characteristics and Definitions Relating to Viruses [00147] Viruses display a wide diversity of shapes and sizes, called 'morphologies'. In general, viruses are much smaller than bacteria. Most viruses that have been studied have a diameter between 20 and 300 nanometers.
  • a complete virus particle consists of nucleic acid surrounded by a protective coat of protein called a capsid.
  • Viruses can have a lipid "envelope" derived from the host cell membrane.
  • the capsid is made from proteins encoded by the viral genome and its shape serves as the basis for morphological distinction. Virally-coded protein subunits will self- assemble to form a capsid, in general requiring the presence of the virus genome.
  • Complex viruses code for proteins that assist in the construction of their capsid. Proteins associated with nucleic acid are known as nucleoproteins, and the association of viral capsid proteins with viral nucleic acid is called a nucleocapsid. The capsid and entire virus structure can be mechanically probed through atomic force microscopy. (Virus, Wikipedia, 2020).
  • Helical Virus - These viruses are composed of a single type of capsomere stacked around a central axis to form a helical structure, which may have a central cavity, or tube. This arrangement results in rod-shaped or filamentous virions which can be short and highly rigid, or long and very flexible.
  • the genetic material typically single-stranded RNA, but ssDNA in some cases
  • the genetic material is bound into the protein helix by interactions between the negatively charged nucleic acid and positive charges on the protein.
  • the length of a helical capsid is related to the length of the nucleic acid contained within it, and the diameter is dependent on the size and arrangement of capsomeres.
  • the well-studied tobacco mosaic virus is an example of a helical virus.
  • Icosahedral Virus Most animal viruses are icosahedral or near-spherical with chiral icosahedral symmetry. A regular icosahedron is the optimum way of forming a closed shell from identical sub-units. The minimum number of identical capsomeres required for each triangular face is 3, which gives 60 for the icosahedron. Many viruses, such as rotavirus, have more than 60 capsomers and appear spherical yet retain symmetry. To achieve this, the capsomeres at the apices are surrounded by five other capsomeres and are called pentons.
  • Virus Envelope Some species of virus envelop themselves in a modified form of one of the cell membranes, either the outer membrane surrounding an infected host cell or internal membranes such as nuclear membrane or endoplasmic reticulum, thus gaining an outer lipid bilayer known as a viral envelope.
  • This tail structure acts like a molecular syringe, attaching to the bacterial host and then injecting the viral genome into the cell.
  • the poxviruses are large, complex viruses that have an unusual morphology.
  • the viral genome is associated with proteins within a central disc structure known as a nucleoid.
  • the nucleoid is surrounded by a membrane and two lateral bodies of unknown function.
  • the virus has an outer envelope with a thick layer of protein studded over its surface.
  • the whole virion is slightly pleomorphic, ranging from ovoid to brick-shaped.
  • Mimivirus is one of the largest characterized viruses, with a capsid diameter of 400 nm. Protein filaments measuring 100 nm project from the surface.
  • the capsid appears hexagonal under an electron microscope, therefore the capsid is probably icosahedral.
  • researchers discovered the largest then known virus in samples of water collected from the ocean floor off the coast of Las Cruces, Chile. Provisionally named Megavirus chilensis, it can be seen with a basic optical microscope.
  • genus Pandoravirus was discovered in Chile and Australia, and has genomes about twice as large as Megavirus and Mimivirus. All giant viruses have dsDNA genomes and they are classified into several families: Mimiviridae, Pithoviridae, Pandoraviridae, Phycodnaviridae, and the Mollivirus genus.
  • a virus has either a DNA or an RNA genome and is called a DNA virus or an RNA virus, respectively. The vast majority of viruses have RNA genomes. Plant viruses tend to have single-stranded RNA genomes and bacteriophages tend to have double-stranded DNA genomes. (Virus, Wikipedia, 2020).
  • Viral genomes are circular, as in the polyomaviruses, or linear, as in the adenoviruses.
  • the type of nucleic acid is irrelevant to the shape of the genome.
  • the genome is often divided up into separate parts, in which case it is called segmented.
  • each segment often codes for only one protein and they are usually found together in one capsid. All segments are not required to be in the same virion for the virus to be infectious, as demonstrated by brome mosaic virus and several other plant viruses. (Virus, Wikipedia, 2020).
  • a viral genome irrespective of nucleic acid type, is almost always either single-stranded or double-stranded.
  • Single-stranded genomes consist of an unpaired nucleic acid, analogous to one-half of a ladder split down the middle.
  • Double-stranded genomes consist of two complementary paired nucleic acids, analogous to a ladder.
  • the virus particles of some virus families, such as those belonging to the Hepadnaviridae contain a genome that is partially double-stranded and partially single-stranded. (Virus, Wikipedia, 2020).
  • the single strands are said to be either positive-sense, called the 'plus-strand', or negative-sense, called the 'minus-strand', depending on if they are complementary to the viral mRNA.
  • Positive-sense viral RNA is in the same sense as viral mRNA and thus at least a part of it can be immediately translated by the host cell. Negative-sense viral RNA is complementary to mRNA and thus must be converted to positive-sense RNA by an RNA-dependent RNA polymerase before translation.
  • DNA nomenclature for viruses with single-sense genomic ssDNA is similar to RNA nomenclature, in that positive-strand viral ssDNA is identical in sequence to the viral mRNA and is thus a coding strand, while negative-strand viral ssDNA is complementary to the viral mRNA and is thus a template strand.
  • ssDNA and ssRNA viruses have genomes that are ambisense in that transcription can occur off both strands in a double- stranded replicative intermediate.
  • examples include Gemini viruses, which are ssDNA plant viruses, and Arenaviruses, which are ssRNA viruses of animals (Virus, Wikipedia, 2020).
  • Genome size varies greatly between species. The smallest—the ssDNA circoviruses, family Circoviridae—code for only two proteins and have a genome size of only two kilobases; the largest—the Pandora viruses—have genome sizes of around two megabases which code for about 2500 proteins. Virus genes rarely have introns and often are arranged in the genome so that they overlap. (Virus, Wikipedia, 2020).
  • RNA viruses have smaller genome sizes than DNA viruses because of a higher error-rate when replicating and have a maximum upper size limit. Beyond this, errors when replicating render the virus useless or uncompetitive. To compensate, RNA viruses often have segmented genomes—the genome is split into smaller molecules—thus reducing the chance that an error in a single-component genome will incapacitate the entire genome. In contrast, DNA viruses generally have larger genomes because of the high fidelity of their replication enzymes. Single-strand DNA viruses are an exception to this rule, as mutation rates for these genomes can approach the extreme of the ssRNA virus case. (Virus, Wikipedia, 2020).
  • RNA viruses often exist as quasi-species or swarms of viruses of the same species but with slightly different genome nucleoside sequences. Such quasi-species are a prime target for studying natural selection and the evolution of replicating RNA, such as from a virus (Fluoreszenzdetetechnische Dictionary molekularer Evolution, Dapprich J, 1994, Cuvillier Verlag Göttingen, ISBN 3-89588-045-0, and: Virus, Wikipedia, 2020).Segmented genomes confer evolutionary advantages; different strains of a virus with a segmented genome can shuffle and combine genes and produce progeny viruses that have unique characteristics. This is called re-assortment or 'viral sex'. (Virus, Wikipedia, 2020).
  • Basic virus life cycle comprises: (1) attachment, (2) penetration, (3) uncoating, (4) replication, (5) assembly, and (6) release.
  • (1) Attachment is a specific binding between viral capsid proteins and specific receptors on the host cellular surface. This specificity determines the host range and type of host cell of a virus. For example, HIV infects a limited range of human leucocytes. This is because its surface protein, gp120, specifically interacts with the CD4 molecule—a chemokine receptor—which is most commonly found on the surface of CD4+ T-Cells. This mechanism has evolved to favor those viruses that infect only cells in which they are capable of replication.
  • Attachment to the receptor can induce the viral envelope protein to undergo changes that result in the fusion of viral and cellular membranes, or changes of non-enveloped virus surface proteins that allow the virus to enter.
  • Virus Wikipedia, 2020.
  • (2) Penetration by plant virus of plant cells, fungal virus penetration of fungal cells, bacterial virus penetration of bacterial cell, and animal virus penetration of animal cells follows viral attachment to these cell types. Virions enter the host cell through receptor-mediated endocytosis or membrane fusion in a process known as viral entry. The infection of plant and fungal cells is different from that of animal cells. Plants have a rigid cell wall made of cellulose, and fungi one of chitin, so most viruses can get inside these cells only after trauma to the cell wall.
  • Viral Uncoating – Uncoating is the process in which the viral capsid is removed: This may be by degradation by viral enzymes or host enzymes or by simple dissociation; the end-result is the releasing of the viral genomic nucleic acid.
  • Viral Replication - Replication of viruses primarily involves multiplication of the genome. Replication involves synthesis of viral mRNA from "early" genes (with exceptions for positive sense RNA viruses), viral protein synthesis, possible assembly of viral proteins, then viral genome replication mediated by early or regulatory protein expression. This may be followed, for complex viruses with larger genomes, by one or more further rounds of mRNA synthesis: "late" gene expression is, in general, of structural or virion proteins. (Virus, Wikipedia, 2020). [00170] (5) Assembly – Following the structure-mediated self-assembly of the virus particles, some modification of the proteins often occurs.
  • viruses such as HIV
  • this modification also referred to as maturation, occurs after the virus has been released from the host cell.
  • Virus Wikipedia, 2020.
  • (6) Release – Viruses can be released from the host cell by lysis, a process that kills the cell by bursting its membrane and cell wall if present: this is a feature of many bacterial and some animal viruses.
  • Some viruses undergo a lysogenic cycle where the viral genome is incorporated by genetic recombination into a specific place in the host's chromosome.
  • the viral genome is then known as a "provirus” or, in the case of bacteriophages a "prophage”. Whenever the host divides, the viral genome is also replicated. The viral genome is mostly silent within the host.
  • the provirus or prophage may give rise to active virus, which may lyse the host cells.
  • Enveloped viruses e.g., HIV
  • HIV enveloped viruses
  • the virus acquires its envelope, which is a modified piece of the host's plasma or other internal membrane.
  • Genome replication The genetic material within virus particles, and the method by which the material is replicated, varies considerably between different types of viruses.
  • DNA viruses The genome replication of most DNA viruses takes place in the nucleus of the cell.
  • RNA viruses - Replication of RNA viruses usually occurs in the cytoplasm. RNA viruses can be placed into four different groups depending on their modes of replication.
  • RNA viruses use their own RNA replicase enzymes to create copies of their genomes. (Virus, Wikipedia, 2020).
  • Reverse transcribing viruses have ssRNA (Retroviridae, Metaviridae, Pseudoviridae) or dsDNA (Caulimoviridae, and Hepadnaviridae) in their particles.
  • Retroviruses integrate the DNA produced by reverse transcription into the host genome as a provirus as a part of the replication process; pararetroviruses do not, although integrated genome copies of especially plant pararetroviruses can give rise to infectious virus. They are susceptible to antiviral drugs that inhibit the reverse transcriptase enzyme, e.g. zidovudine and lamivudine.
  • HIV which is a retrovirus
  • Hepadnaviridae which includes Hepatitis B virus.
  • Virus Wikipedia, 2020.
  • Cytopathic effects on the host cell The range of structural and biochemical effects that viruses have on the host cell is extensive. These are called 'cytopathic effects'. Most virus infections eventually result in the death of the host cell. The causes of death include cell lysis, alterations to the cell's surface membrane, and apoptosis. Often cell death is caused by cessation of its normal activities because of suppression by virus-specific proteins, not all of which are components of the virus particle. The distinction between cytopathic and harmless is gradual.
  • viruses such as Epstein– Barr virus
  • papillomaviruses are established causes of cancer.
  • Virus, Wikipedia, 2020 [00177] Dormant and latent infections - Some viruses cause no apparent changes to the infected cell. Cells in which the virus is latent and inactive show few signs of infection and often function normally. This causes persistent infections and the virus is often dormant for many months or years. This is often the case with herpes viruses. (Virus, Wikipedia, 2020).
  • Host range - Viruses are by far the most d biological entities on Earth, outnumbering all the others put together.
  • viruses infect all types of cellular life including animals, plants, bacteria, and fungi. Different types of viruses can infect only a limited range of hosts and many are species-specific. Some, such as smallpox virus, can infect only one species, in this case humans, and are said to have a narrow host range. Other viruses, such as rabies virus, can infect different species of mammals and are said to have a broad range. The viruses that infect plants are harmless to animals, and most viruses that infect other animals are harmless to humans. The host range of some bacteriophages is limited to a single strain of bacteria, which can be used to trace the source of outbreaks of infections by a method called phage typing.
  • nucleic acids shall mean sequences of nucleic acids containing a few nucleotides to thousands of nucleotides, wherein the nucleic acids further may comprise a single or multiple polymers in a sample and may comprise double stranded nucleic acids conformations or single strand nucleic acids conformations.
  • Biological samples generally contain a large number of different enzymes which can rapidly degrade nucleic acids in various ways., Worldwide during the COVID-19 pandemic, available methods for detecting SARS-CoV-2 are problematic and do not provide the methods and compositions of the present invention in numerous ways, including biological sample collection guidelines for medical technicians, researchers, hospital staff, nurses, and doctors do not provide the lysis buffer compositions of the present invention.
  • SARS-CoV-2 sampling and testing of the sample should use conventional methods including: (1) a conventional method of collecting biological matter from a person and placing the biological matter immediately into several (1 to 3 ml) milliliters of a CDC VTM (Virus Transfer Medium) buffer or an aqueous phosphate buffered saline (PBS); and (2) then a conventional method of using the VTM or PBS buffer for a period of days to weeks to maintain a state of freshness in the biological samples.
  • CDC VTM Virus Transfer Medium
  • PBS aqueous phosphate buffered saline
  • This conventional method unwittingly maintains a potential for virus particles, if present in the biological sample, to be capable of known viral activities such as inadvertent infection of workers handling the sample or transmission to other samples resulting in cross-contamination and reduced accuracy.
  • Such uncontrolled viral activity is a potential direct health hazard for personnel collecting, storing, transporting and disposing of samples and presents substantial risks especially to those working with the samples in a laboratory setting and doing the actual testing.
  • This conventional method also maintains the risk of unnecessarily allowing potentially rapid degradation of nonhuman nucleic acids (DNA and/or RNA) in the biological sample prior to testing for the presence of virus particles or nucleic acids to be tested.
  • the present invention uses a lysis buffer to inactivate the nucleic acid hydrolyzing enzymes by quickly rendering and keeping these proteins in denatured inactive conformations, thereby increasing the accuracy of test results.
  • a lysis buffer to inactivate the nucleic acid hydrolyzing enzymes by quickly rendering and keeping these proteins in denatured inactive conformations, thereby increasing the accuracy of test results.
  • some embodiments of the present invention practice a method including obtaining a biological sample from a selected source of biological matter; contacting the biological sample without delay with an amount of a lysis buffer(s) to form a mixture(s), wherein the lysis buffer comprises a protein denaturing agent in a fluid, and using the protein denaturing agent(s) from the fluid lysis buffer in the mixture as a means for storing the mixture for a time period of storage or transporting the mixture over a period of time, which may be several weeks or 1-6 weeks, before testing, analyzing, and/or identifying the biological matter source of identified non-human nucleic acids.
  • RNA and/or DNA identification and nucleic acids include NGS (next generation sequencing) including robotic machine microprocessor controlled processing of multiple test samples using RT-PCR, LAMP-PCR, and many other PCR methods which utilize a variety of optical multiplexing methods with one or more optical dyes as probes, TaqMan and various selected polynucleotide primers for specific identification of non-human nucleic acids sequences for viruses such as SARS-CoV-2, bacteria, pathogenic life forms, and microorganisms.
  • NGS next generation sequencing
  • a protein denaturing agent may comprise a high concentration amount of an aqueous chaotropic salt(s) or an organic compound base or organic compound salt such as urea and/or an alkylated urea(s) or a guanidine salt(s) or a derivatized guanidine compound base(s) or/and guanidine salt(s).
  • the protein denaturing agent(s) may be used to cause an immediate lysis of biological cells, microorganisms, bacteria, virus particles, and any other infectious organisms that may have come from the biological matter in the biological sample.
  • This lysis activity can be used to help sterilize the biological matter in the biological sample without delay after it is obtained from a human, an animal, or another source/location such as a place or surface which a human may use.
  • Denaturing protein enzymes in the biological sample are one means for inactivating the nuclease enzymes (RNases, DNases) which are capable of rapidly degrading nucleic acids in the biological sample.
  • RNases, DNases nuclease enzymes
  • Agents which can be used to denature proteins include chaotropic agents and disulfide bond reducers.
  • Chaotropic agents include: Urea of 6 – 8 mol/L, guanidine hydrchloride 6 mol/L, Lithium perchlorate 4.5 mol/L, and Sodium dodecyl sulfate __ mol/L.
  • Disulfide bond reducers include the following chemicals: 2-Mercaptoethanol, Dithiothreitol, TCEP (tris(2-carboxyethyl)phosphine).
  • Guanidine exists protonated, as guanidine, in solution at physiological pH.
  • Guanidinium chloride also known as guanidine hydrochloride
  • guanidine hydrochloride In aqueous solutions containing 6 M guanidine hydrochloride, almost all proteins lose their entire secondary structure and become randomly coiled peptide chains.
  • Guanidine thiocyanate is also used for its denaturing effect on various biological samples.
  • Guanidine exists protonated, as guanidine, in solution at physiological pH.
  • the guanidine core structure groups R1, R2, R32, R4, and R5 may be selected from the group consisting of hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, 2-pentyl, 3-pentyl, isopentyl, tert-pentyl, straight chain alkyl C6-C10 groups, branched chain alkyl C6-C10 groups, straight chain alkene C1-C10 groups, branched chain alkene C1-C10 groups, C5-C8 cycloalkyl groups, C5-C8 cycloalkenyl groups, C5-C8 cycloaryl groups, polyhexamethylene, and any combination thereof.
  • the central bond within this group is that of an imine, and the group is related structurally to amidines and ureas.
  • Chaotropic Anions -
  • the guanidine organic cation or another chaotropic cation in a lysis buffer embodiment of the present invention is accompanied by a chaotropic anion.
  • a chaotropic anion may be selected from the group consisting of chloride, nitrate, bromide, iodide, thiocyanate, chlorate, acetate, fluoride, chlorite, thiocyanate, phosphate, hydrogen phosphate, dihydrogen phosphate, propionate, butyrate, nitrite, benzoate, sulfite, sulfide, sulfate, sulfonate, formate, methanoate, glycerate, citrate, malate, malonate, urate, succinate, oxalate, carbonate, bicarbonate, thiophosphate, ferrocyanide, pyrophosphate, and a combination thereof.
  • the guanidine salt for the lysis buffer can be selected from the group consisting of guanidine hydrochloride, guanidine chloride, guanidine nitrate, guanidine bromide, guanidine iodide, guanidine thiocyanate, guanidine chlorate, guanidine acetate, guanidine sulfate, guanidine fluoride, guanidine chlorite, guanidine thiocyanate, guanidine phosphate, guanidine hydrogen phosphate, guanidine dihydrogen phosphate, guanidine propionate, guanidine butyrate, guanidine nitrite, guanidine benzoate, guanidine sulfite, guanidine sulfide, guanidine formate, guanidine methanoate, guanidine glyceride, guanidine citrate, guanidine mal
  • Typical lysis buffer embodiments of the present invention contain one or more different anions in concentrations between 0.01 molar to 5 molar.
  • the lysis buffer contains a mixture of anions wherein each anion is present in an amount less than 4 molar, more preferably wherein each anion is present in an amount less than 2.5 molar.
  • Some lysis buffer embodiments of the present invention have an anion selected from the group consisting of a hydrochloride, a hydrobromide, a hydroiodide, a sulfate, a nitrate, a hydrogen phosphate, a phosphate, an acetate, a propionate, a hexanoate, cyclopentanepropionate, glycolate, a pyruvate, a lactate, malonate, a succinate, maliate, maleate, fumarate, tartrate, a citrate, benzoate, 3-(4-hydroxybenzoyl) benzoate, cinnaminate, mandelate, methanesulfonate, a besylate, an ethanesulfonate, 1,2- ethanedisulfonate, 2-hydroxyethane-sulfonate, benzenesulfonate, 4- chlorobenzenesulfonate, 2-naphthalenes
  • Typical lysis buffer embodiments of the present invention contain one or more different cations in concentrations between 0.01 molar to 5 molar.
  • the lysis buffer contains a mixture of cations wherein each anion is present in an amount less than 4 molar, more preferably wherein each cation is present in an amount less than 2.5 molar.
  • the cation has a concentration of less than 0.5 molar.
  • Some lysis buffer embodiments of the present invention have a cation selected from the group consisting of aluminum ion, ammonium ion, antimony ion, arsenic ion, barium ion, beryllium ion, bismuth ion, boron ion, bromide ion, cadmium ion, calcium ion, cerium ion, cesium cation, chloride ion, chromium ion, cobalt ion, copper ion, dysprosium ion, erbium ion, europium ion, fluoride ion, gadolinium ion, gallium ion, germanium ion, gold ion, hafnium ion, holmium ion, indium ion, iodine ion, iridium ion, iron ion, lanthanum ion, lead ion, lithium ion, lute
  • Some lysis buffer embodiments comprise at least one inorganic salt with a cation which is not an organic cations having a nitrogen atom, but which is an inorganic cation selected from the group consisting of cesium, lithium, sodium, potassium, iron, zinc, manganese, cobalt, nickel, and copper.
  • Invention Embodiments of Lysis Buffer Comprise a Protein Denaturing Agent
  • the lysis buffer may comprise a high concentration of an aqueous chaotropic salt solution for some embodiments of the present invention, wherein the concentration is between about 1 to about 10 molar; about 4 to about 8 molar, or about 4 to about 6 molar.
  • the buffer needs to be prepared using water free of ribonucleases (RNases).
  • RNases ribonucleases
  • examples of RNase-free water may be selected from the group consisting of a medical grade water, a molecular biology grade water, a water treated by reverse osmosis, a double distillation water, an ultrafiltration water, a sterilized water, a membrane filtration water, a charcoal filtered water, a water used for the preparation of injections, a renal dialysis water, an intravenous use water, and any combination thereof.
  • the RNase-free water may be purified using an column comprising RNA covalently linked to a solid substrate such as beads as a means for lowering RNase levels in the water to be used to make the buffer.
  • a solid substrate such as beads
  • an immobilized RNA-containing column or disk can be attached to a syringe containing a water sample to push through the RNA-containing column so as to remove RNases from the water.
  • Example composition using an example lysis buffer [00204]
  • the buffer may comprise an RNase-free water and between about 2 M and 6 M guanidine thiocyanate.
  • the buffer may further comprise for example a concentration of sodium citrate between about 1 mM to about 100 mM with a pH 6-7.
  • the buffer may further comprise from 0.1 mM to about 100 mM EDTA to help inhibit DNases if DNA is also being extracted for its identification.
  • a silica-based spin column Silica-based spin columns for the isolation of nucleic acids from biological samples are commercially available for example from: Qiagen, ThermoFisher, Zymo, Macherey Nagel.
  • a collection tube also referred to as microcentrifuge tube or Eppendorf tube. Silica-based spin columns and collection tubes are frequently supplied together in a single kit.
  • a specimen swab can be used that comprises a top portion (the handle) that can be broken off so that the swab portion of the specimen swab fits completely into a sample collection tube, thus allowing the sample collection tube to be fully closed with a cap or other type of seal, such as a rubber stop. Perform a specimen collection from the patient or individual being sampled using the specimen swab, such as described previously in the specification.
  • lysis buffer also termed “RELY” buffer “RELY SAFE” buffer, or “RELY” can be used in downstream testing assays in its entirety, thereby allowing for an approximately 4x (four-fold) increase in detection sensitivity compared to protocols where the biological matter is diluted into a fluid, such as VTM, UTM or PBS, in order to prepare a biological sample for testing or analysis.
  • Adding the biological matter specimen directly and immediately into a lysis buffer makes the SARS-CoV-2 nucleic acids assay more sensitive for RNA detection, which reduces false negative results - enables earlier detection.
  • Examples of lower detection limits when using the lysis buffer of the present invention are about 1 – 100 virions per mL.
  • An example lysis buffer embodiment of the present invention to prepare the biological sample mixture is depicted in FIG.2C.
  • a lysis buffer requiring (1) a very high concentration of a chaotropic salt(s) or urea as a denaturing agent, and (2) a disulfide reducing agent as a lysis composition suitable to inactivate ribonuclease in nucleic acids containing biological matter specimens.
  • a disulfide reducing agent as a lysis composition suitable to inactivate ribonuclease in nucleic acids containing biological matter specimens.
  • Currently available compositions include the disulfide reducing agent, to avoid rapid enzymatic degradation of the nucleic acids by ribonucleases in the biological matter or sample specimen.
  • Embodiments of the present invention are effective lysis buffer compositions without a reducing agent.
  • a surprising finding of the present invention is that that omitting the beta-mercaptoethanol disulfide reducing agent did not matter! Even without a disulfide reducing agent, the lysis buffer was still fully effective in lysing cells, denaturing proteins and protecting nucleic acids from ribonucleases.
  • the present invention lysis buffer embodiments have the advantage in lacking a disulfide reducing agent because reducing agents such as beta-mercaptoethanol are odorous, volatile, unstable (must be bought fresh frequently) and potentially toxic to human use.
  • disulfide bond reducers examples include 2-Mercaptoethanol, Dithiothreitol (DTT), TCEP (tris-2-carboxyethyl phosphine), dithiobutyl-amine (DTBA), and a combination thereof.
  • DTT Dithiothreitol
  • TCEP tris-2-carboxyethyl phosphine
  • DTBA dithiobutyl-amine
  • RNase R ribonuclease peptide inhibitors
  • reducing agents such as DTT and B- mercaptoethanol which disrupt disulphide bonds in the RNase enzyme, but the effect is limited and temporary as well as being toxic and volatile
  • proteases such as proteinase K to digest the RNases, but the transport of proteinases in kits and their generally slow action allows the analyte biomolecules to degrade.
  • Bates (2011) also states at paragraph [0011]: “Traditionally RNA degradation is avoided by keeping the contact time between the guanidine and the lysate containing the RNA to a minimum; sample lysis is generally immediately followed by separation of the RNA from the guanidine.
  • PCR methods are continuously being challenged and improved.
  • the CDC published a CDC 2019-Novel Coronavirus (2019-nCoV) Real-Time RT-PCR Diagnostic Panel which is a real-time RT-PCR test intended for the qualitative detection of nucleic acid from the 2019-nCoV in upper and lower respiratory specimens (such as nasopharyngeal or oropharyngeal swabs, sputum, lower respiratory tract aspirates, bronchoalveolar lavage, and nasopharyngeal wash/aspirate or nasal aspirate) collected from individuals who meet 2019-nCoV clinical and/or epidemiological criteria (for example, clinical signs and symptoms associated with 2019-nCoV infection, contact with a probable or confirmed 2019-nCoV case, history of travel to geographic locations where 2019-nCoV cases
  • CDC 2019-nCoV Real-Time RT-PCR Diagnostic Panel is a molecular in vitro diagnostic test that aids in the detection and diagnosis 2019-nCoV and is based on widely used nucleic acid amplification technology.
  • the product contains oligonucleotide primers and dual-labeled hydrolysis probes (TaqMan®) and control material used in rRT-PCR for the in vitro qualitative detection of 2019-nCoV RNA in respiratory specimens.
  • the oligonucleotide primers and probes for detection of 2019- nCoV were selected from regions of the virus nucleocapsid (N) gene.
  • the panel is designed for specific detection of the 2019-nCoV (two primer/probe sets).
  • An additional primer/probe set to detect the human RNase P gene (RP) in control samples and clinical specimens is also included in the panel.
  • the CDC procedure is to isolate RNA and purify RNA from upper and lower respiratory specimens.
  • RNA is then reverse transcribed to cDNA and subsequently amplified in the Applied Biosystems 7500 Fast Dx Real-Time PCR Instrument with SDS version 1.4 software.
  • the probe anneals to a specific target sequence located between the forward and reverse primers.
  • the 5’ nuclease activity of Taq polymerase degrades the probe, causing the reporter dye to separate from the quencher dye, generating a fluorescent signal.
  • additional reporter dye molecules are cleaved from their respective probes, increasing the fluorescence intensity. Fluorescence intensity is monitored at each PCR cycle by Applied Biosystems 7500 Fast Dx Real-Time PCR System with SDS version 1.4 software.
  • RNA is generally detectable in upper and lower respiratory specimens during infection. Positive results are indicative of active infection with 2019-nCoV but do not rule out bacterial infection or co-infection with other viruses. The agent detected may not be the definite cause of disease. Laboratories within the United States and its territories are required to report all positive results to the appropriate public health authorities. The CDC states that negative results do not preclude 2019-nCoV infection and should not be used as the sole basis for treatment or other patient management decisions. Negative results must be combined with clinical observations, patient history, and epidemiological information.
  • SARS-CoV-2 virus SARS coronavirus
  • RT-PCR reverse transcriptase polymerase chain reaction assays
  • RNA and DNA are aqueous solutions of only the salts disodium hydrogen phosphate, sodium chloride, potassium chloride, and potassium dihydrogen phosphate (Sigma Aldrich, St. Louis, MO).
  • Step 2 Swirl the swab vigorously for 15 seconds, then expunge as much liquid as possible from the swab by pressing and rotating the fiber portion against the wall of the tube. Discard the swab [00221] Step 2. To each sample, add 10 ⁇ l GXP Mag Beads and mix well by pipetting up and down. [00222] Step 3. Incubate the tube for 5 min at 65°C or omit this step if for certain the material at this step contains no active virus. Proteinase K can work better with a small amount of detergent, may not be relevant here [00223] Step 4. Transfer the tube onto the magnetic rack and magnetize the samples for 5 min or until the solution appears clear. [00224] Step 5.
  • Step 6 Add 200 ⁇ l Ethanol (80%) to each tube. Do not mix! Incubate for 30 sec. Remove and discard the supernatant without disturbing the bead pellet.
  • Step 7. Remove and discard the supernatant without disturbing the bead pellet.
  • Step 8. Add 200 ⁇ l Ethanol (80%) to each tube. Do not mix! Incubate for 30 sec. Remove and discard the supernatant without disturbing the bead pellet. Verify the presence of residual ethanol! [00228] Step 9. Remove the tube from the magnet and air dry the pellet for 5-10 min. [00229] Step 10.
  • Step 11 Incubate the tube for 5 min.
  • Step 12. Transfer the tube onto the magnetic rack and incubate the samples for 3-5 min or until the solution appears clear.
  • Step 13 Aspirate 50 ⁇ l of the supernatant to a new tube.
  • the eluted RNA/DNA can be used immediately for molecular based applications or stored at 80oC for future use.
  • the mineral substrate used for adsorbing a nucleic acid molecule is preferably a filter that comprises or consists of porous or non-porous metal oxides or mixed metal oxides, silica gel, sand, diatomaceous earth, materials predominantly consisting of glass, such as unmodified glass particles, powdered glass, quartz, alumina, zeolites, titanium dioxide, and zirconium dioxide.
  • Fiber filters comprised of glass and any other material that can be molded into a fiber filter may be employed in this method.
  • alkaline earth metals are used in the mineral substrate, they may be bound by ethylenediaminetetraacetic acid (EDTA) or EGTA, and a sarcosinate may be used as a wetting, washing, or dispersing agent. Any of the materials used for the mineral substrate may also be engineered to have magnetic properties.
  • the particle size of the mineral substrate is preferably from 0.1 um to 1000 um, and the pore size is preferably from 2 to 1000 um.
  • the mineral substrate may be found loose, in filter layers made of glass, quartz, or ceramics, in membranes in which silica gel is arranged, in particles, in fibers, in fabrics of quartz and glass wool, in latex particles, or in frit materials such as polyethylene, polypropylene, and polyvinylidene fluoride especially ultra-high molecular weight polyethylene, high density polyethylene.”
  • the heat stable Taq (‘AmpliTaq’) enzyme has a 5’-exonucleolytic function that does not displace the probe sitting in the middle but instead digests it, releasing the reporter dye in the process.
  • Phi29 is a polymerase that is not heat stable (40C max) but which has a strand displacement function.
  • FIG.14 depicts aspects of the TaqMan kit and process and SYBR Green, and reagents used to detect PCR products.
  • the addition of sulfate ions are used to improve the non-infectiousness of the lysis buffer composition and reagent solution for increased safety.
  • the lysis buffer composition here called ‘RELY’, provides up to 10x more sensitive testing compared to other collection buffers, such as viral transport medium (VTM), universal transport medium (UTM), physiological or normal saline solution, ‘salina’ buffer solution, phosphate buffered saline (PBS), and others.
  • VTM viral transport medium
  • UDM universal transport medium
  • PBS phosphate buffered saline
  • Example 5 Sensitivity testing of RELY Lysis Buffer compared to PBS
  • Direct PCR (without the use of a separate RNA extraction step) was carried out by diluting the lysate sample 1:100 with nuclease-free water. 1 ⁇ l of the 1:100 diluted lysate sample was used for qPCR and run in triplicate after adding 19 ⁇ l PCR Mastermix with a Primer/Probe mix for CoV-2 genes N1 and M (RNA) und the human gene RNase P (DNA) as a control.
  • the volumes used in this control test are considered standard for typical SARS-CoV-2 collection media (4 ml PBS, UTM or VTM), in contrast to a significantly (10x) reduced volume that to be used with RELY (400 ⁇ l).
  • GuSCN guanidine thiocyanate, guanidine thiocyanate or guanidine thiocyanate
  • GITC guanidine thiocyanate
  • a solution of 4.5M GuHCl + 0.45M K 2 SO 4 was used.
  • a solution of 4.5M GuSCN + 0.45M K 2 SO 4 was used.
  • a solution of 4.5M GuHCl + 0.45M Na 2 SO 4 is used.
  • a solution of 4.5M GuSCN + 0.45M Na 2 SO 4 is used.
  • a solution of 4.5M GuHCl + 0.45M K 2 SO 4 is buffered with sodium acetate (NaCH3COO, also abbreviated NaOAc) to obtain pH 5-6.
  • NaOAc sodium acetate
  • a solution of 4.5M GuSCN + 0.45M K 2 SO 4 is buffered with NaOAc to obtain pH 5-6.
  • a solution of 4.5M GuHCl + 0.45M Na 2 SO 4 is buffered with NaOAc to obtain pH 5-6.
  • a solution of 4.5M GuSCN + 0.45M Na 2 SO 4 is buffered with NaOAc to obtain pH 5-6.
  • a solution of 4.5M GuHCl + 0.45M K 2 SO 4 is buffered with Tris (tris(hydroxymethyl)aminomethane, (HOCH2)3CNH2) to obtain pH 7-8.5.
  • Tris tris(hydroxymethyl)aminomethane, (HOCH2)3CNH2)
  • a solution of 4.5M GuSCN + 0.45M K 2 SO 4 is buffered with Tris to obtain pH 7-8.5.
  • a solution of 4.5M GuHCl + 0.45M Na 2 SO 4 is buffered with Tris to obtain pH 7-8.5.
  • a solution of 4.5M GuSCN + 0.45M Na 2 SO 4 is buffered with Tris to obtain pH 7-8.5.
  • a solution of 4.5M GuHCl + 0.45M K 2 SO 4 is buffered with sodium citrate to obtain pH 5-6.
  • Sodium citrate can refer to any of the three sodium salts of citric acid. One of these sodium salts of citric acid, trisodium citrate, has the chemical formula Na3C6H5O7. Another of these sodium salts of citric acid, trisodium citrate, has the chemical formula Na 2 C 6 H 6 O 7 . A combination of the two can be used to arrive at pH 5-6. Alternatively a solution of HCl (hydrochloric acid) or NaOH (sodium hydroxide) can be used to adjust pH. An example for how to obtain a buffered Tris or TE solution is given below.
  • a solution of 4.5M GuSCN + 0.45M K 2 SO 4 is buffered with sodium citrate to obtain pH 5-6.
  • a solution of 4.5M GuHCl + 0.45M Na 2 SO 4 is buffered with sodium citrate to obtain pH 5-6.
  • a solution of 4.5M GuSCN + 0.45M Na 2 SO 4 is buffered sodium citrate to obtain pH 5-6.
  • Tris Solution 1. Dissolve 121 g Tris base in 800 ml H2O. 2. Adjust to desired pH with concentrated HCl. Approximately 70 ml HCl is needed to achieve a pH 7.4 solution, and 42 ml for a pH 8.0 solution. 3.
  • FIG.15 shows typical commercially used volumes of viral transport or collection media (4000 ⁇ l of UTM, VTM, PBS, salina) were compared to a typical volume of RELY (400 ⁇ l).
  • RELY was prepared as 4.5M GuHCl + 0.45M K 2 SO 4 buffered with 1M NaOAc for pH 5 and the N1 viral gene of SARS-CoV-2 was tested in qPCR, comparing the relative sensitivity of RELY vs PBS. Results were run in triplicate and are shown in the table below as “Rep1”, “Rep2”, “Rep3” as well as the Mean value of all three. We get a 3.7 times better CT (cycle threshold qPCR) value for 0.4 ml RELY versus 4 ml PBS. In other words, RELY is 12.6 times more sensitive than other typically used types and volumes of viral collection and transport media and viral lysis buffers.
  • the present RELY buffer has a functional component (sodium sulfate, potassium sulfate, sodium citrate, Tris, NaOAc) that gives it better COVID RNase and human RNase protein insolubility.
  • the RELY buffer has an alkaline pH 8-8.5 that gives the SARS-CoV-2 sample less infectivity immediately after collection from a patient.
  • the RELY buffer has an acidic pH 5-6 that gives the SARS-CoV-2 sample less infectivity immediately after collection from a patient.
  • the RELY buffer contains one ingredient selected from the group of guanidine hydrochloride, guanidine thiocyanate, guanidine sulfate; contains no reducing agent, and contains an additional different function component selected from the group of sodium sulfate, potassium sulfate, sodium citrate, Tris or NaOAc, and any combination thereof.
  • a 4.5M GuHCl + 0.45M K 2 SO 4 buffered with 1M NaOAc at pH 5 (column #3, in bold) was generated by adding 90 ⁇ l of a stock solution of 5M GuHCl and 0.5M K 2 SO 4 , 5 ⁇ l of 1 M NaOAc at pH 5, 2.5 ⁇ l of SARS-CoV-2 reference (Zeptometrix 1:100), and 2.5 ⁇ l RNase P plasmid (IDT 1:1) for a total volume of 100 ⁇ l.
  • FIG.17 is a bar graph providing a data overview of the qPCR results for the SARS-CoV-2 reference RNA gene ‘N1’.
  • FIG.18 is a bar graph providing a data overview of the qPCR results for the SARS-CoV-2 reference RNA gene ‘M’.
  • a data overview for the SARS-CoV-2 reference RNA gene ‘M’ is given below:
  • FIG.19 is a bar graph providing a data overview of the qPCR results for the human control DNA gene RNase P (‘RP’).
  • RP RNase P
  • FIG.20 is a bar graph with PCR cycles on left axis needed for COV-2 Genes N1, M1, and RP tested using a RELY Buffer formulation No.3.
  • FIG.21 is a bar graph showing a data overview for the SARS-CoV-2 three reference genes ‘N1’, ‘M’ and ‘RP’ as tested in PBS.
  • BM Volume per Reaction Buffer Mix
  • EM Enzyme Mix
  • PPM Primer / Probe Mix
  • CTR Positiv Control
  • Nuclease-free Water 2.8 Total Reaction Volume 20 Step Temp Time Cycle RT 50°C 10min 1 Initial Denaturation 95°C 2min 1 Denaturation 95°C 5sec 50 Anneal/Elongation 60°C 30sec 50
  • BM Volume per Reaction Buffer Mix
  • EM Enzyme Mix
  • PPM Primer / Probe Mix
  • CTR Positiv Control
  • Nuclease-free Water 2.8 Total Reaction Volume 20 Step Temp Time Cycle RT 50°C 10min 1 Initial Denaturation 95°C 2min 1 Denaturation 95°C 5sec 50 Anneal/Elongation 60°C 30sec 50
  • Additional Lysis Buffer Compositions Embodiments of the Invention: [00280] It is contemplated that the lysis buffer is optimized to solve several potential needs. [00281] Example 7.
  • a lysis buffer consists of an aqueous composition comprising at least 3 ingredients in a volume of a ribonuclease (RNase) free sterile water: [00283] (1) a Cesium salt having a concentration of 0.010 – 4 Molar, of 0.050 – 4 Molar, preferably of 0.1-3 Molar, of 0.1-2 Molar, of 0.1-1 Molar, of 0.1-0.5 Molar; and most preferably of 0.15 to 0.3 Molar; [00284] (2) a pH buffer having a concentration of 0.025-200 millimolar with the pH adjusted to [00285] pH 6-7, to pH 5.9-7.5, and preferably to between pH 6.8-7.1.
  • RNase ribonuclease
  • the pH buffer may be sodium citrate, sodium hydrogen phosphate, a Tris Buffer, a “Good” Buffer or another pH buffer.
  • the pH may be adjusted to the desired pH using a base such as sodium hydroxide or potassium hydroxide or any other basic chemical; and [00286] (3) a Guanidine salt having a concentration of between about 5-6 Molar, between about 4-5 Molar, between about 3-4 Molar, between about 2-3 Molar, between about 1-2 Molar, between about 0.5-1 Molar, between about 0.2-0.5 Molar, between about 0.1-0.3 Molar.
  • the guanidine salt is preferably guanidine hydrochloride or guanidine thiocyanate.
  • a multivalent ion chelator such as EDTA, EGTA, NTA, HEDTA and the like may be included having a concentration between about 0.005 to 0.2 Molar.
  • a lysis buffer comprises an aqueous composition comprising at least 3 ingredients in a volume of a ribonuclease (RNase) free sterile water: [00290] (1) a Sodium salt or a potassium salt or a mixture of them having a concentration between about 0.1 – 4 Molar, 0.150 – 3 Molar, between about preferably 0.15 -2 Molar, between about 0.15 -0.1 Molar, between about 0.2-0.5 Molar, between about 0.2-0.3 Molar, more preferably 0.15 to 0.25 Molar.
  • RNase ribonuclease
  • a pH buffer having between about 0.010-.200 Molar pH buffer set to pH 6- 7, between about pH 5.9-7.5, preferably between about pH 6.6-7.2, more preferably between about 6.9-7.1.
  • the pH buffer may be sodium citrate, sodium hydrogen phosphate, a Tris Buffer, a “Good” Buffer or another pH buffer.
  • the pH may be adjusted to the desired pH using a base such as sodium hydroxide or potassium hydroxide; and [00292] (3) a Guanidine salt having a concentration between about 5-6 Molar, between about 4-5 Molar, between about 3-4 Molar, between about 2-3 Molar, between about 1-2 Molar, between about 0.5-1 Molar, between about 0.2-0.5 Molar, between about 0.1-0.3 Molar.
  • the guanidine salt is preferably guanidine hydrochloride or guanidine thiocyanate.
  • a multivalent ion chelator such as EDTA, EGTA, NTA, HEDTA and the like may be includes having a concentration between about 0.005 to 0.2 Molar.
  • a lysis buffer comprises an aqueous composition comprising at least 3 ingredients in a volume of a ribonuclease (RNase) free sterile water: [00296] (1) a Lithium salt having a concentration 0.1 – 4 Molar, 0.150 – 3 Molar, preferably 0.15 -2 Molar, 0.15 -0.1 Molar, 0.2-0.5 Molar, 0.2-0.3 Molar, more preferably 0.15 to 0.25 Molar. Sodium sulfate and/or potassium sulfate are preferred.
  • a pH buffer having 0.025-200 millimolar of the pH buffer set to pH 6-7, pH 5.9-7.5, preferably to between pH 6.8-7.1.
  • the pH buffer may be sodium citrate, sodium hydrogen phosphate, a Tris Buffer, a “Good” Buffer or another pH buffer.
  • the pH may be adjusted to the desired pH using a base such as sodium hydroxide or potassium hydroxide; and
  • a Guanidine salt having a concentration 5-6 Molar, 4-5 Molar, 3-4 Molar, 2-3 Molar, 1-2 Molar, 0.5-1 Molar, 0.2-0.5 Molar, 0.1-0.3 Molar.
  • the guanidine salt is preferably guanidine hydrochloride or guanidine thiocyanate.
  • a multivalent ion chelators such as EDTA, EGTA, NTA, HEDTA and the like may be includes having a concentration between about 0.005 to 0.2 Molar.
  • a lysis buffer comprises an aqueous composition [00302] comprising at least 3 ingredients in a volume of a ribonuclease (RNase) free sterile water: [00303] (1) Sodium Sulfate salt having a concentration between about 0.05 – 0.2 Molar, 0.150 – 0.3 Molar, preferably 0.15 -0.3 Molar, 0.15 -0.1 Molar, 0.2-0.5 Molar, 0.2-0.3 Molar, more preferably 0.15 to 0.25 Molar. In one embodiment 200 millimolar sodium sulfate salt is used.
  • RNase ribonuclease
  • a pH buffer having 0.010-200 millimolar of the pH buffer set for example to pH 6.0, 6.2, 6.4, 6.6, 6.8, 7.0, 6-7, pH 5.9-7.5, pH 6.8-7.1.
  • the pH buffer may be 25 millimolar sodium citrate buffer, a sodium hydrogen phosphate buffer , a Tris Buffer, a “Good” Buffer or any other pH buffer.
  • the pH may be adjusted using a base such as sodium hydroxide or potassium hydroxide; and
  • a Guanidine salt having a concentration 5-6 Molar, 4-5 Molar, 3-4 Molar, 2-3 Molar, 1-2 Molar, 0.5-1 Molar, 0.2-0.5 Molar, 0.1-0.3 Molar.
  • the guanidine salt is 5 molar guanidine hydrochloride or 5 molar guanidine thiocyanate.
  • a Cesium salt may be selected from the group consisting Cesium bromide, Cesium chloride, Cesium iodide, Cesium fluoride, Cesium sulfide, Cesium selenide, Cesium carbonate, Cesium chromate, Cesium dichromate, Cesium dihydrogen phosphate, Cesium bicarbonate, Cesium bisulfate, Cesium monohydrogen phosphate, Cesium nitrate, Cesium nitrite, Cesium perchlorate, Cesium permanganate, Cesium phosphate, Cesium sulfate, Cesium sulfite, Cesium thiosulfate, Cesium silicate, Cesium metasilicate, Cesium aluminum silicate, Cesium acetate, Cesium formate, Cesium oxalate, Cesium methanoate, Cesium glycerate, Cesium
  • a Lithium salt may be selected from the group consisting Lithium bromide, Lithium chloride, Lithium iodide, Lithium fluoride, Lithium sulfide, Lithium selenide, Lithium carbonate, Lithium chromate, Lithium dichromate, Lithium dihydrogen phosphate, Lithium bicarbonate, Lithium bisulfate, Lithium monohydrogen phosphate, Lithium nitrate, Lithium nitrite, Lithium perchlorate, Lithium permanganate, Lithium phosphate, Lithium sulfate, Lithium sulfite, Lithium thiosulfate, Lithium silicate, Lithium metasilicate, Lithium aluminum silicate, Lithium acetate, Lithium formate, Lithium oxalate, Lithium methanoate, Lithium glycerate, Lithium citrate, Lithium citrate, Li
  • a Potassium salt may be selected from the group consisting of Potassium bromide, Potassium chloride, Potassium iodide, Potassium fluoride, Potassium sulfide, Potassium selenide, Potassium carbonate, Potassium chromate, Potassium dichromate, Potassium dihydrogen phosphate, Potassium bicarbonate, Potassium bisulfate, Potassium monohydrogen phosphate, Potassium nitrate, Potassium nitrite, Potassium perchlorate, Potassium permanganate, Potassium phosphate, Potassium sulfate, Potassium sulfite, Potassium thiosulfate, Potassium silicate, Potassium metasilicate, Potassium aluminum silicate, Potassium acetate, Potassium formate, Potassium oxalate, Potassium methanoate, Potassium glycerate, Potassium citrate, Potassium citrate,
  • a Sodium salt may be selected from the group consisting of Sodium bromide, Sodium chloride, Sodium, Sodium fluoride, Sodium sulfide, Sodium selenide, Sodium carbonate, Sodium chromate, Sodium dichromate, Sodium dihydrogen phosphate, Sodium bicarbonate, Sodium bisulfate, Sodium monohydrogen phosphate, Sodium nitrate, Sodium nitrite, Sodium perchlorate, Sodium permanganate, Sodium phosphate, Sodium sulfate, Sodium sulfite, Sodium thiosulfate, Sodium silicate, Sodium metasilicate, Sodium aluminum silicate, Sodium acetate, Sodium formate, Sodium oxalate, Sodium methanoate, Sodium glycerate, Sodium citrate, Sodium malate, Sodium malonate, Sodium urate, Sodium succinate, Sodium oxalate, Sodium sulfonate
  • Example 11 The table below shows Storage Stability Testing conducted with the variable of storage temperature on how well RELY Lysis Buffer Compared to PBS Buffer acts to protect COV-2 Zeptometrix gene N1 and COV-2 Zeptrometrix gene M1. Three temperatures were tested: RT(room temperature approximately 20 ° C), 35 ° C and 50 ° C. These temperatures can easily occur in parts of the world without air conditioning such as during storage in a truck. [00312] Amplification plots are created when the fluorescent signal from each sample is plotted against cycle number; therefore, amplification plots represent the accumulation of product over the duration of the real-time PCR experiment.
  • the Rn value is the fluorescent signal from SYBR Green normalized to (divided by) the signal of the passive reference dye for a given reaction.
  • the delta Rn value is the Rn value of an experimental reaction minus the Rn value of the baseline signal generated by the instrument. This parameter reliably calculates the magnitude of the specific signal generated from a given set of PCR conditions.
  • FIG.22 is a graph of storage stability testing conducted with variable storage temperatures on showing how well RELY Lysis Buffer Compared to PBS Buffer acts to protect COV-2 Zeptometrix gene N1.
  • FIG.23 Is a graph of Storage Stability Testing conducted at variable storage temperatures showing how well RELY Lysis Buffer Compared to PBS Buffer acts to protect COV-2 Zeptometrix gene M1.
  • Three temperatures were tested: RT(room temperature approximately 20 ° C), 35 ° C and 50 ° C. These temperatures can easily occur in parts of the world without air conditioning such as during storage in a truck.
  • the temperature resistant effectiveness of RELY Buffer means full protection of RNA at 20 ° C, 35 ° C, and 50 ° C.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Analytical Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Plant Pathology (AREA)
  • Virology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

This disclosure provides compositions and methods useful for detecting the presence of viral ribonucleic acid (RNA) in a biological sample. Compositions and methods are provided for SARS-CoV-2 virus testing.

Description

LYSIS BUFFER COMPOSITIONS AND METHODS FOR PREPARING A VIRAL BIOLOGICAL SAMPLE USEFUL FOR COVID-19 TESTING CROSS-REFERENCE TO RELATED APPLICATIONS [0001] This application claims the benefit of U.S. Provisional Application No.63/103,435 filed on August 5, 2020, and U.S. Provisional Application No.63/205,592 filed on December 24, 2020 each of which are herein incorporated by reference in their entirety. FIELD OF THE INVENTION [0002] The present invention relates to methods and lysis buffer compositions useful for preparing a sample specimen, such as a viral biological sample, for refrigeration-free shipping, storage, and more sensitive, accurate, rapid, safe, SARS-CoV-2 virus testing. The present invention is also useful for testing for other human RNA viruses or pathogens. SARS-CoV-2 stands for “severe acute respiratory syndrome coronavirus 2”, the virus responsible for causing the coronavirus disease “COVID-19”. SARS- CoV-2 was previously known as “2019 novel coronavirus”. Many documents, information sites, guidelines and testing protocols use the two terms SARS-CoV-2 and COVID-19 somewhat interchangeably but typically refer to testing procedures for the presence of SARS-CoV-2 in individuals that may develop COVID-19 symptoms. BACKGROUND OF THE INVENTION [0003] The COVID-19 virus pandemic which has infected more than 16 million individuals and causing more than 650,000 fatalities since its 2019 outbreak. A major concern of the public and health officials (Center For Disease Control (CDC) and World Health Organization (WHO)) has been that the SARS-CoV-2 virus (SARS coronavirus) is more stable than most viruses in fluid samples and in human environmental surfaces and may be capable of persisting long enough to cause airborne viral transmission. [ [0004] SARS-CoV-2 infection sites include lower respiratory tract infections, pneumonia, and fatal acute respiratory distress syndrome (ARDS) and infections in other organs. SARS-CoV-2 infection causes inflammatory diseases which worsen in patients already having lung diseases such as asthma, chronic obstructive pulmonary disease, or emphysema and such patients often need intensive medical care. Elderly and immune diseased persons are at higher risk that young adults. Severe COVID-19 related disease is fatal due to pneumonia-related progressive respiratory failure (Emami, 2020). [0005] Both false negative and false positive results are suspected to be occurring in SARS-CoV-2 tests at rates between 5-40 % depending on sample collection, storage, instrumentation, methodology, and assays used during testing (Kucirka LM et al., Variation in false-negative rate of reverse transcriptase polymerase chain reaction– based SARS-CoV-2 Tests by time since exposure, Annals of Internal Medicine, May 13, 2020; Brody J, The case for smarter coronavirus testing, The New York Times, July 13, 2020; FDA warns of false positives with BD coronavirus diagnostic, FDA Brief, Taylor NP, July 7, 2020). The so-called e-gene specific primers of the WHO real-time PCR assay have been reported to generate up to 10% false positives, which have been found to result at least in part from a partial match of the e-gene specific primers intended to identify SARS-CoV-2 to a 70 bp long region that matches a bacterial species common in the nasopharyngeal environment, Neisseria subflava. An improved real-time PCR assay with higher specificity and potentially lower rates of false positives, called the M-Gene Assay, has since been developed (Optimized RT- PCR Approach for the Detection of Intra- and Extra-Cellular SARS-CoV-2 RNAs, International Journal of Molecular Sciences, Toptan T et al., June 20, 2020). [0006] Controlling the spread of COVID-19 by quarantining individuals who are infected is difficult because the symptoms of this infection can be hidden for 3-14 days; during this time an infected person is infectious and can infect many other people with the SARS-CoV-2 virus. The CDC-sanctioned SARS-CoV-2 test has a significant percentage of false negative test results. Early research indicates that the common test for SARS-CoV-2 produces false negative test results up to 30% of the time (‘False Negatives’ in COVID-19 Testing: If You Have Symptoms, Assume You Have the Disease, Pratt, 2020, internet publication source: https://www.healthline.com/health- news/false-negatives-covid19-tests-symptoms-assume-you-have-illness). [0007] The major concern for false negative test results is that many asymptomatic people who test negative for SARS-CoV-2 virus will believe that they are not infected and therefore unknowingly spread the virus into their community. Conversely, people who test positive for SARS-CoV-2 virus may believe that they are now immune to COVID-19, but they may still be susceptible to the diseases and its transmission to others if the test was a false positive. This situation has led to an urgent need for a rapid, safe, and accurate SARS-CoV-2 test method to determine if a person is infected with the SARS-CoV-2 virus, but this need has not been met. [0008] A large COVID-19 study indicated a vast (6-24 fold) undercounting of SARS- CoV-2 infection cases (Seroprevalence of Antibodies to SARS-CoV-2 in 10 Sites in the United States, March 23-May 12, 2020, JAMA Internal Medicine, Havers et al., July 22, 2020). This means the number of people creating the risk of SARS-CoV-2 infection (based on SARS-CoV-2 antibody detection) is higher than the risk of SARS- CoV-2 infection indicated by the number of people with COVID-19 symptoms. This disparity means there is a very urgent need for a rapid, safe, and accurate SARS-CoV-2 test method to determine if a person is infected with the SARS-CoV-2 virus but this need has not been met. For example, Dr. Elizabeth Rosenthal in the New York Times on July 24, 2020 reported that COVID-19 testing can take two weeks from the time of obtaining a biological sample from a person to the time the report is issued to the person. Waiting two weeks for test results makes this test, in this case, pointless, since SARS-CoV-2 infection symptoms take 3 to 14 days to appear. Having to wait too long for SARS-CoV-2 test results makes early quarantining not possible and does not aid in slowing the growth of the COVID-19 pandemic. The terms “biological sample”, “biological specimen”, “sample specimen” as used here are the same as the term “biological matter”. [0009] SARS-CoV-2 test methods have at least one unneeded dilution step. and need better safety measures in place. SARS-CoV-2 test methods need to be less sensitive to ambient temperatures during shipping and storage. Existing SARS-CoV-2 test methods are inconvenient in that they require refrigeration of the sample specimen to protect test accuracy and sensitivity. Currently available SARS-CoV-2 test methods are not efficient and rapidly deployed and tested, which causes shocking public realization that there are still significant backlogs, uncertainties and inadequacies in testing for SARS- CoV-2 infection in humans. Delays in SARS-CoV-2 testing of up to two weeks have been reported and to some extent are due to using test methods, which are methodologically defective in their procedural expediency. Even more problematic is that delaying the testing of a biological sample for traces of nucleic acids is fraught with false negative test results. SARS-CoV-2 tests may have faulty designs or incorrectly adapted biological sample preparation methods allowing for significant biological sample deterioration and release of ribonucleases and deoxy-ribonucleases from cells in the biological sample, which causes rapid and significant degradation of the nucleic acids sequences of the SARS-CoV-2 virus in the biological sample needed to accurately detect if the biological specimen from a human shows that the human is infected with SARS-CoV-2 virus. These are some of the problems which embodiments of the present invention have overcome, particularly the potential low accuracy of the prior art SARS-CoV-2 tests, which can cause false negative test results. [0010] Furthermore, it is known that laboratory tests are characterized by their ability to detect a positive case (sensitivity) and their ability to detect a negative case (specificity). Sensitive tests are less likely to provide a false-negative result and specific tests are less likely to provide a false-positive result. SARS-CoV-2 test manufacturers and laboratories often report the “analytic” sensitivity and specificity of their test based on analyses of highly artificial sets of known positive and negative samples for marketing and sales purposes that demonstrate the highest potential accuracy of the SARS-CoV-2 tests under highest ideal conditions including biological samples very recently collected from patients with high viral loads or from patients with a complete absence of SARS-CoV-2 exposure. Sensitivity and specificity of testing based on real-world conditions of patients and their biological samples is highly important. For example, when a patient’s biological sample is improperly collected or the RNA in the biological sample has degraded, most certainly a SARS-CoV-2 tested person will be erroneously told they are not infected with SARS-CoV-2. [0011] Published on March 13, 2020 by the U.S. CDC in response to the Coronavirus 2019 outbreak is a CDC SOP #DSR-052-03 entitled: Preparation of Viral Transport Medium (VTM) in 3 ml screw cap tubes. The VTM contains 2% fetal bovine serum, Hanks Balanced Salt Solution with calcium and magnesium ions, and Gentamycin sulfate, Amphotericin B to be stored at 2-8oC (Relich for CDC, 2020). The CDC’s VTM is explicitly for transport of biological specimens. Human biological samples from persons who may have SARS-CoV-2 infection are collected and diluted in titer in a volume of VTM and are shipped. [0012] In May 22, 2020, the CDC Issued Interim Guidelines for Collecting, Handling, and Testing Clinical Specimens for COVID-19 Specimen Collection: https://www.cdc.gov/corona virus/2019-nCoV/lab/guidelines-clinical-specimens.html. [0013] The term Viral Transport Medium (VTM) is often used somewhat interchangeably with the term Universal Transport Medium™ (UTM®), a room temperature stable viral transport medium for collection, transport, maintenance and long term freezer storage of virus specimens. [0014] Collection and testing protocols for biological samples often include the need to refrigerate the samples. A major SARS-CoV-2 virus testing lab company, Quest Diagnostics, issued guidelines on June 29, 2020 which include refrigerating biological samples in dry ice that are made ready for transporting and storage prior to their testing. A refrigeration step is not practical for sample collections of the biological matter, such as in the case of virus detection testing of humans living under poor conditions, in wilderness areas, or countries with limited capabilities and resources. It is an unresolved problem in the that human biological samples must be frozen during their transport to centers where the samples can be tested for the presence of SARS- CoV-2 virus test. While freezing has been long known to inactivate nucleases and infectivity in, for example, virus-containing biological samples, it has become a modern world habit and requirement. The inactivation-by-freezing method however is not only an inconvenience for countries and settings that lack such capabilities in their routine operations: the widespread inability to carry out inactivation-by-freezing presents a significant health and life hazard and contributes greatly to inaccurate downstream test results. Furthermore, there is need for safety from viruses in biological test samples as well, so that a person testing the viral sample does not accidently become infected by the active virus in prior art biological samples, as there is always some risk of accidental exposure during viral sample shipping, storage, or testing for the virus that can expose a person to active virus from a biological sample. The terms ‘biological sample’, ‘biological specimen’, ‘sample specimen’ and ‘biological matter’ are used interchangeably herein. [0015] The test kits used on SARS-CoV-2 virus samples need to be accurate despite the fact that the original SARS-CoV-2 virus genome has been found capable of appearing with thousands of random mutations. There are doubts that these SARS-CoV-2 virus test kits are capable of being sufficiently accurate given the number of already known and newly emerging mutations. For example, the SARS-CoV-2 mutation known as D614G mutant may greatly matter for infectivity and disease progression (Schraer, 7- 19-2020 BBC News “Coronavirus: Are mutations making it more infectious?”). D614G mutated SARS-CoV-2 virus has a virus spike protein change which increases infectivity of SARS-CoV-2 virus to humans. Needed are test kits which can accurately detect D614G and other mutations of the SARS-CoV-2 genome to stay aware of the pandemic evolution of SARS-CoV-2 virus genome. [0016] A dependence of SARS-CoV2 infectivity and disease progression to a person’s HLA and MHC type has been observed (Is there a genetic link to COVID-19 disease severity? Free T., BioTechniques, July 27, 2020). Gene-based risks related to COVID- 19 overlay the currently known risks such as age and pre-existing conditions and are governed primarily by a region on chromosome 6 called HLA/MHC. This region is responsible for the immune system’s defense against pathogens. Evolution has made this region extremely diverse so that - as a species - humans do not get wiped out by a newly emerged pathogen such as the one presented by the current pandemic. Individuals, however, can have very different genetic risk profiles. Studies of COVID- 19 urgently look for information on gene-based risk of infection, disease progression and risk of death. Understanding the correlations among these factors can lead to personalized interventions that can help better protect vulnerable groups and save patients’ lives through specific treatments. Perhaps more importantly, the development of personalized treatments and vaccines may be improved and sped up through patient stratification based on correlated information about a patient’s SARS-CoV-2 predominant genetic variant(s), their genotype and expression profile, and their COVID-19 disease progression [0017] Biological samples from many biological matter nucleic acid sources, such as a person, will comprise biological fluids, cells, cell molecules, and various enzymes, in addition to the biological matter nucleic acid source infectious containing pathogens such as a virus. The nuclease enzymes in a biological sample can rapidly destroy the RNA or DNA in the biological sample after this sample is obtained and before the testing to detect the RNA or DNA is implemented. There are nuclease enzymes in nearly all biological samples. [0018] Nuclease enzymes can hydrolyze nucleic acid phosphodiester bonds and can chemically alter nucleic acid bases. Ribonucleases specifically degrade RNA. Deoxyribonucleases specifically degrade DNA. Non-specific nucleases degrade both RNA and DNA. Polynucleotide phosphorylase and pyrophosphorylase, are also capable of depolymerizing RNA. [0019] Phosphomonoesterases bind to polynucleotides or oligonucleotides at a terminal phosphate group or mononucleotide to remove a phosphate group. Endonucleases produce oligonucleotides which thereby cause a lowering of the viscosity of the nucleic acid as well as decreasing the ability of a test kit to identify the nucleic acid virus. Exonucleases release mononucleotides from nucleic acids. Some nucleases are both endonucleases and exonucleases. The third feature is mode of phosphodiester bond cleavage. Polynucleotides can be cleaved at the phosphodiester bond on either side of the phosphate. Of the ribonucleases, those enzymes that produce 3' -phosphates and 5' –hydroxyl termini are concerned with the degradation of RNA whereas those enzymes that are involved in the processing of RNA precursor molecules generate 5' - phosphate and 3' -hydroxyl termini. Other distinguishing features of nucleases can be made based on their specificity towards secondary structures, directions of attack of the polynucleotide, and maximum enzymatic nuclease velocity. [0020] Also, nucleic acids may be modified by telomerase, also called terminal transferase, a ribonucleoprotein that adds a species-dependent telomere repeat sequence to the 3' end of a telomere, which is a region of repetitive sequences at each end of eukaryotic chromosomes (in most eukaryotes) to protect the end of the chromosome from DNA damage or from fusion with neighboring chromosomes. Also, nucleic acids may be modified by a transposable element (transposon, or jumping gene), which is a DNA sequence that can change its position within a genome, sometimes creating or reversing mutations and altering the cell's genetic identity and genome size. Transposition often results in duplication of the same genetic material. There are at least two classes of transposons: Class I transposons or retrotransposons generally function via reverse transcription, while Class II transposons or DNA transposons encode the protein transposase, which they require for insertion and excision of DNA [0021] Also, nucleic acids may be modified by methyl transferase enzymes, a class of enzymes which can methylate two of DNA's four bases, cytosine and adenine. Adenine or cytosine methylation is part of the restriction modification system of many bacteria. Cytosine methylation is widespread in both eukaryotes and prokaryotes, even though the rate of cytosine DNA methylation can differ greatly between species. Adenine methylation has been observed in bacterial, plant, and mammalian DNA. [0022] In addition, nucleic acids may be modified by helicases, which are a class of enzymes that move along a nucleic acid phosphodiester backbone, separating two annealed nucleic acid strands such as DNA and RNA. Many cellular processes, such as DNA replication, transcription, translation, recombination, DNA repair, and ribosome biogenesis involve the separation of nucleic acid strands that necessitates the use of helicases. Helicases may create nucleic acid fragments. Thus, it is a problem that there are a number of nucleic acids altering and degrading enzymes in typical biological samples. These enzymes are ubiquitous, capable of being released from cells, not inactivated by fluids used to collect the biological matter to make a biological sample, and will lower the original amount of viral or microbial RNA or DNA in the biological sample, thereby leading to a lack of sensitivity in the test. Consequently, these virus detection kits suffer a risk of low accuracy by causing false negative test results. Primarily, it is believed that biological samples for detection testing of a virus RNA instantly begin to degrade. RNA virus detection test kits use a buffer to immediately inactivate at least ambient ribonuclease (RNase) enzyme activity in the biological sample. Low sensitivity test kits for nucleic acids have the problem of false negative results because low levels of nucleic acids in a test sample occur when the test kit procedure uses a buffer which excessively dilutes a test sample and then uses only a fraction of the diluted test sample to detect nucleic acids. [0023] One way to inactivate enzymes which destroy RNA and DNA is to use a process of enzyme protein denaturation, commonly by using 5-10 molar urea or a guanidine salt. Urea in high concentrations disrupts the noncovalent bonds in the proteins. Guanidine hydrochloride (also known as guanidinium chloride) has chaotropic properties which denature proteins. A chaotropic agent is a substance which disrupts the structure of, and denatures, macromolecules such as proteins and nucleic acids (e.g. DNA and RNA). Chaotropic solutes increase the entropy of the system by interfering with intermolecular interactions mediated by non-covalent forces such as hydrogen bonds, van der Waals forces, and hydrophobic effects. In aqueous solutions containing 6M guanidine chloride, almost all proteins lose their entire secondary structure and become randomly coiled peptide chains. Guanidine thiocyanate is also used for its denaturing effect on various biological samples (Guanidine, Wikipedia, 2020). [0024] Currently available virus detection test kits based on detecting nucleic acids include a reducing agent such as about a 0.1 molar concentration of 2-mercaptoethanol (also known as β-mercaptoethanol, BME, 2BME, 2-ME, β-met, and HOCH2CH2SH) to reduce disulfide bonds to irreversibly denature the ribonuclease enzymes in biological samples being tested for the presence of a virus. However, use of such reducing agents is problematic..A reducing agent such as2-Mercaptoethanol is used in RNA isolation procedures to eliminate ribonuclease released during cell lysis by adding it to the sample storage buffer to guarantee the denaturation of the ribonuclease. The mechanism understood by which proteins are denatured by β-mercaptoethanol involves a cleavage of the disulfide bonds that may form between thiol groups of cysteine residues in a protein. Of course, 0.1 molar is a large excess compared to a biological fluid sample. Excess 2-mercaptoethanol shifts the equilibrium reaction rightward: RS– SR + 2 HOCH2CH2SH ⇌ 2 RSH + HOCH2CH2S–SCH2CH2OH. The protein symbolized by RS-SR with a –S-S- disulfide bond bridging two chains of a protein is reduced to form 2 RSH with the result that the protein chains are unlinked at a critical region needed for ribonuclease activity. [0025] There is a need for compositions for detecting viral RNA in a biological sample that does not require unnecessary dilution of the biological sample, does not include unstable and undesirable reducing agents, increased sensitivity, and lower risk of infectivity to healthcare workers and laboratory technicians handling and testing the biological samples. SUMMARY OF THE INVENTION [0026] Provided herein are compositions for immediately and efficiently creating surprisingly stable biological samples not needing a reducing agent or refrigeration. The  compositions are useful for detecting the presence of viral ribonucleic acid (RNA) in a biological samples. The compositions herein include a lysis buffer having a protein denaturing agent wherein the protein denaturing agent inactivates proteins in the mixture, prevents degradation of nucleic acids, sterilizes the biological sample without the need for refrigeration and wherein the lysis buffer does not include a reducing agent. [0027] The compositions protect nucleic acids initially present in the biological sample as a part of the specimen collection process by causing an inhibition of the degradation of the nucleic acids until the biological sample mixture can be tested for a pathogen such as SARS-CoV-2 virus. This inhibition is achieved by including a protein denaturing agent in a lysis buffer of the composition The compositions can be immediately prepared anywhere rapidly and efficiently and safely as the biological matter is being collected. [0028] In another embodiment, provided herein are methods for detecting a viral ribonucleic acid (RNA) in a biological sample contacting the biological sample with a lysis buffer including a protein denaturing agent to form a mixture wherein the protein denaturing agent inactivates proteins in the mixture, prevents degradation of nucleic acids, and sterilizes the biological sample, and wherein the lysis buffer does not include a reducing agent and the method does not require unneeded dilution of the nucleic acids into a fluid, such as VTM, UTM or PBS before contacting the biological sample with a composition of the present invention. [0029] Over dilution of the biological sample lowers test sensitivity and accuracy particularly if the person’s biological specimen is problematic in having low virus titer to start with. [0030] In one aspect, the compositions and methods of the present invention lessen viral infection risk to a virus sample handler or tester: The lysis buffer is needed to reduce rate of binding or affinity of a coronavirus to a bind and infect a mammalian cell. This would help to make the lysis buffer safer for the person taking the viral sample from that person if contacted then becoming infected with the virus on their body anywhere. This is done by selecting a pH which is not conducive to coronavirus attachment to a cell or replication in a mammalian cell should one remain intact. [0031] In another aspect, the denaturing agent in the lysis buffer of the compositions described herein inactivates the virus by denaturing the virus proteins. [0032] The compositions of the present invention stabilize the virus RNA from enzymes such as a ribonuclease (RNase). In one embodiment, the compositions include one or more cations or anions that bind to the viral RNA and inhibit its degradation by an RNase. [0033] In another embodiment, hydrolysis of viral RNA is reduced or minimized by controlling the pH of the composition. [0034] In another embodiment the compositions of the present invention avoid the use of a reducing agent. Surprisingly, the elimination of reducing agents in the compositions and methods of the present invention does not affect the accuracy of the viral test results. Even without a disulfide reducing agent, the lysis buffer of the compositions herein are fully effective in lysing cells, denaturing proteins and protecting nucleic acids from ribonucleases, and avoid undesirable characteristics of the reducing agents which are odorous, volatile, unstable (must be bought fresh frequently) and potentially toxic to human use. [0035] In another embodiment, the concentration of guanidine salt in the lysis buffer of the compositions is minimized to increase the stability of the composition. [0036] In one embodiment, the lysis buffer has a functional component such as, for example, sodium sulfate, potassium sulfate, sodium citrate, Tris, and/or NaOAc) to provide better COVID RNase and human RNase protein insolubility and minimize degradation of viral RNA in a biological sample. BRIEF DESCRIPTION OF THE DRAWINGS [0037] FIGS.1A-1C show a method of biological sample preparation using a flat surface to which is first added biological matter to be lysed by spraying a lysis buffer on the biological matter to form biological sample mixture. [0038] FIGS.2A-2C show a method of biological sample mixture preparation using a collection tube in which a lysis buffer is first added to the collection tube and then a biological matter is added to the collection tube to be lysed and to form the biological sample mixture. [0039] FIGS.3A-3C depict a method of biological sample mixture preparation using a flat surface to which is first added a lysis buffer, and then added is biological matter to be lysed so as to form a biological sample mixture [0040] FIGS.4A-4C depict a procedure wherein a wet nucleic acids sample is added to a porous surface which may be a filter surface of any kind capable of retaining nucleic aci [0041] FIG.5A depicts a procedure wherein a collection tube has contents which are a biological sample comprising a lysis buffer fluid with denatured proteins, lysed cell proteins, lipids, and nucleic acids, and optionally further comprising an added nucleic acid. The contents of the sample collection tube 501 comprise a lysis buffer fluid with sample specimen 502, comprising denatured proteins 510 and cellular debris, lysed proteins and lipids 510, and with nucleic acids to be tested 503, and optionally further comprising added nucleic acids 505. [0042] FIG.5B depicts performing a non-magnetic isolation of the nucleic acid to be tested 503 by running a spin column 507 having a spin column material 509 in a collection tube 508. Added nucleic acids 505 may be retained along with nucleic acids to be tested 503 in the spin column material 509 or washed and discarded into the flow-through 506 materials which include denatured proteins 510 and cellular debris, lysed proteins and lipids 510. [0043] FIG.5C depicts the nucleic acids to be tested 503 and optional added nucleic acid 505 in nuclease-free water 511. [0044] FIG.5D depicts an example spin column procedure for purifying RNA and DNA from collection buffer mixture using ethanol for extracting the RNA and DNA and washing the nucleic acids free of the lysis buffer and denatured proteins and cellular debris. [0045] FIGS.6A-6D depict a procedure for a magnetic bead-based purification of nucleic acids RNA and DNA from oral swab collected biological matter for SARS-CoV-2 virus. [0046] FIGS.7A-7D depict purification of nucleic acids in a mixture of collected biological matter containing a lysis buffer through use of a centrifugal filter membrane. [0047] FIGS.8A-8C depict a reverse transcriptase PCR (RT-PCR) process. [0048] FIG.9 depicts in accordance with some embodiments of the present invention, several common kinds of biological matter (sample specimen) collection devices. [0049] FIGS.10A-10E depict a sequencing procedure to identify nucleic acid mutations for samples giving a positive nucleic acid test result. [0050] FIG.11 depicts an overview of the steps in Next Generation Sequencing workflow. [0051] FIG.12 is a schematic of steps in the workflow of a next generation sequencing based assay. [0052] FIG.13 depicts some of the steps which may be used for some embodiments of the present invention for collecting cellular biological matter specimen in a collection tube, lysing cells in the cellular biological matter specimen in the collection tube, binding RNA from the collected cellular biological matter specimen to a column with a material which binds RNA, and washing and eluting the RNA from the RNA binding column into a tube for holding the RNA in a fluid. [0053] FIG.14 depicts aspects of the TaqMan Kit and Process and SYBR Green 1 Reagents used to detect PCR products [0054] FIG.15 is a graph showing validation data of the sensitivity of a RELY lysis buffer of the present invention vs PBS (phosphate buffered saline). [0055] FIG.16 is a bar graph with PCR Cycles needed on left axis to begin to detect COV-2 Gene N1. [0056] FIG.17 is a bar graph providing a data overview of the qPCR results for the SARS-CoV-2 reference RNA gene ‘N1’. [0057] FIG.18 is a bar graph providing a data overview of the qPCR results for the SARS-CoV-2 reference RNA gene ‘M’. [0058] FIG.19 is a bar graph providing a data overview of the qPCR results for the human control DNA gene RNase P (‘RP’). [0059] FIG.20 is a bar graph with PCR cycles on left axis needed for COV-2 Genes N1, M1, and RP tested using a RELY Buffer formulation No.3. [0060] FIG.21 is a bar graph showing a data overview for the SARS-CoV-2 three reference genes ‘N1’, ‘M’ and ‘RP’ as tested in PBS. [0061] FIG.22 is a graph of storage stability testing conducted with variable storage temperatures on showing how well RELY Lysis Buffer Compared to PBS Buffer acts to protect COV-2 Zeptometrix gene N1. Three temperatures were tested: RT(room temperature approximately 20°C), 35°C and 50°C. [0062] FIG.23 is a graph of storage stability testing conducted with variable storage temperatures on showing how well RELY Lysis Buffer Compared to PBS Buffer acts to protect COV-2 Zeptometrix gene M1. Three temperatures were tested: RT(room temperature approximately 20°C), 35°C and 50°C. DETAILED DESCRIPTION OF THE INVENTION [0063] General embodiments of the present invention comprise testing methods and test kits including immediate use lysis buffer(s) useful for detecting and making various qualitative and quantitative assessments of the presence of virus, bacterial, fungal, animal, and plant biological matter in a test sample. Using methods and test kits and buffer compositions of the present invention, nucleic acids may be obtained from a nucleic acids source selected from the group consisting of a virus, a bacteria, a fungus, an animal, a human, a plant, a lab sample, an environment, a dwelling, a public place, a food sample, a water sample, an air sample, a soil, a synthetic nucleic acids source, and any combination thereof. A nucleic acids sample from biological matter or lab sample is a particularly important source for testing during the ongoing SARS-CoV-2 virus pandemic. Using methods and test kits and buffer compositions of the present invention, a test sample containing nucleic acids may be safely, rapidly, efficiently, accurately detected with high sensitivity, and the nucleic acids may accurately be characterized as a means for identifying a biological source of the nucleic acids. A test kit for detecting SARS-CoV-2 virus nucleic acids needs to safely address obstacles to proper nucleic acids detection including the problem of ambient nucleic acids enzymes in biological samples. [0064] Currently available compositions use a biological sample collection fluid comprising a CDC-authorized VTM (Virus Transfer Medium), PBS (phosphate buffered saline), Amies transport medium, sterile saline, and a combination thereof during initial biological matter collection, however this is unnecessary for practicing the many embodiments of the present invention. Using the currently existing biological sample collection fluid is responsible for reported SARS-CoV-2 virus test false negative results for several reasons. Collections of, for example, an upper respiratory specimen such as, for example, a nasopharyngeal (NP) specimen, an oropharyngeal (OP) specimen, a nasal mid-turbinate swab, an anterior nares or nasopharyngeal wash/aspirate, or nasal wash/aspirate (NW) specimen from a person who may have become infected with a SARS-CoV-2 virus is a needless dilution of the nucleic acids, the source of which may include nucleic acids from a nucleic acids source selected from the group consisting of a virus, a bacteria, a fungus, an animal, a human, a plant, a lab sample, an environment, a dwelling, a public place, a food sample, a water sample, an air sample, a soil sample, a synthetic nucleic acids source, and any combination thereof. In general, collections of samples which may be a source of nucleic acids selected from the group consisting of a virus, a bacteria, a fungus, an animal, a human, a plant, a lab sample, an environment, a dwelling, a public place, a food sample, a water sample, an air sample, a soil sample, a synthetic nucleic acids source, and any combination thereof. This dilution step is a significant problem that can cause false negative test results in methods attempting to detect the nucleic acids. [0065] Obtaining a Biological Sample from a Selected Source of Biological Matter [0066] For initial diagnostic testing for SARS-CoV-2, CDC recommends a healthcare worker use a swab to collect an upper respiratory specimen such as, for example, a nasopharyngeal (NP) specimen, an oropharyngeal (OP) specimen, a nasal mid- turbinate swab, an anterior nares or nasopharyngeal wash/aspirate or nasal wash/aspirate (NW) specimen from a person who may have become infected with a SARS-CoV-2 virus. The CDC recommends that the upper respiratory swab specimen should be placed immediately into a sterile transport tube containing 2-3mL of either viral transport medium (VTM), Amies transport medium, or sterile saline. Testing lower respiratory tract specimens is also an option. For patients who develop a productive cough, sputum should be collected and tested for SARS-CoV-2. The induction of sputum is not recommended. When under certain clinical circumstances (e.g., those receiving invasive mechanical ventilation), a lower respiratory tract aspirate or broncho-alveolar lavage sample should be collected and tested as a lower respiratory tract specimen. The CDC teaches that other swab specimens, such as originating from tongue or saliva, have decreased sensitivity and are unacceptable, and indicates that more data are necessary to better understand the validity of sample specimens from buccal swabs, saliva, or other specimen types for SARS-CoV-2 testing. Saliva tests allow a patient to spit into a tube rather than have their nose or throat swabbed. Saliva tests may be more comfortable and less invasive for test subjects and are likely safer for healthcare workers due to increased distance during sample collection. [0067] A non-magnetic process embodiment of the present invention can be used to test for the presence of the virus in an oral swab biological sample taken from a person suspected of being infected with the virus. The sample can be taken from the nose, the mouth, the eyes, the throat, a cut, a wound, from sputum, vomit, excrement, urine, blood, semen, sweat, tears, lymph fluid, or other bodily fluids. The swab can collect a sample from under the tongue, the throat, an oral location, or another region in the body which may be infected or contaminated with the virus. A sample does not necessarily have to originate from a swab, it can also be collected from saliva, spit, sputum, a syringe needle biopsy or from a sample obtained from the body of a deceased person. The reagent composition of the present invention or the lysis buffer may be applied onto a wall or another surface of a support material (such as paper) or container (such as a collection tube) in liquid or dry form, or be applied for instance as a spray. [0068] The present invention method embodiments and compositions may comprise using a lyophilized lysis buffer composition that stabilizes the biological sample upon contact, such as within 15-30 seconds or more generally 5-60 seconds after allowing a person to spit into a dry collection tube that comprises the lysis buffer. One advantage of the present invention is that even in dried form (when the concentration reagent composition is higher than in a preferred `liquid embodiment as described), it does not harm the biological composition preparation or the downstream testing and analysis performed after sample collection and optional storage and shipping. [0069] The invention embodiments provide various compositions comprising a lysis buffer and may further comprise a salt. The lysis buffer may be immobilized on a tissue, paper, tube, or swab. In one invention embodiment as a method, a person may cough or spit on invention embodiments which provide the various compositions of the invention which comprise a lysis buffer and may further comprise a salt, wherein the lysis buffer and or lysis salts are present as a solid, a gel, a damp area, a fluid, or in a releasable formulation immobilized or loosely present on or wetting for example a tissue, a paper, a tube or a swab which function as a method using a kit comprising a sampling device or sample receptacle providing a lysis means for constituents comprising the biological matter which may contain an amount of an infectious pathogen such as a virus such as SARS-CoV-2. Such a receptacle for a biological specimen or sample due to the presence of a lysis buffer embodiment of the present invention can easily be shipped without having to refrigerate the receptacle for a biological specimen or sample. Alternatively, lysis medium or even dry lysis composition from a foil or plastic container could be added immediately to the sample container or collection tube, receptacle, or biological sample soon after a person provided the specimen of biological matter or the lysis composition may be delivered as a spray onto the biological specimen which may contain the infectious pathogen such as SARS-CoV-2. [0070] For some embodiments of the present invention the practicing methods and compositions of the lysis buffer may further comprise an added nucleic acid optionally comprising a unique genetic sequence that are included in the biological sample or lysis buffer composition used. An added nucleic acid is a term for a chemical marker which can be added and used as a method embodiment of the present invention comprising: an internal control for verifying correct, accurate and safe operation of the sample collection and testing procedure, tracking the biological sample during biological sample preparation; shipping or transporting the biological sample; testing the biological sample; reporting results of the biological sample; and organizing results of various tests of the biological sample by same or multiple labs, and facilitating tested persons’ HIPAA (Health Insurance Portability and Accountability Act of 1996) rights to anonymous protection of the tested persons health test data, and safe dissemination of the test data in a publication or for demographics regarding the COVID-19 pandemic. [0071] In addition, some of the embodiments of the present invention are useful for testing for a virus on a surface of equipment, instrumentation, tools, protective equipment, intubation tubes, masks, surfaces, clothing, active working or living environments, air filters in heating and vacuum systems, hospital morgues, doctor’s office, TSA, police departments, or immigration offices. [0072] It is advantageous that some lysis buffer embodiments of the present invention can be prepared and stored over weeks, months, or years before it is being used. [0073] Virus test kits use biological samples which detect virus nucleic acids, RNA (ribonucleic acid), or DNA (deoxyribonucleic acid) depending on the target virus or microorganism. RNA and DNA are polymers composed of nucleotide monomers which each have three components: a 5-carbon sugar, a phosphate group, and a nitrogenous base. The sugar in an RNA nucleotide is ribose and is deoxyribose in a DNA nucleotide. Strings of nucleotides are bonded to form helical backbones - typically, one for RNA, two for DNA - and assembled into chains of base-pairs selected from the five primary, or canonical, nucleobases: adenine, cytosine, guanine, thymine, and uracil. Thymine occurs only in DNA and uracil only in RNA. The sugars and phosphates in nucleic acids are connected to each other in an alternating chain (sugar-phosphate backbone) through phosphodiester linkages. In conventional models, the carbons to which the phosphate groups attach are the 3'-end and the 5'-end carbons of the sugar. Ends of nucleic acid molecules are called the 5'-end and the 3'-end. The nitrogenous bases (nucleobases) are bonded to the sugars via an N-glycosidic linkage between a nucleobase ring nitrogen (N-1 for pyrimidines and N-9 for purines) and the 1' carbon of the sugar. Cell RNA (ribonucleic acid) comes in three universal forms: transfer RNA (tRNA), messenger RNA (mRNA), and ribosomal RNA (rRNA). There are also non-standard nucleosides in both RNA and DNA usually having arisen from a modification of the nucleosides when in a DNA molecule or a primary RNA transcript of the DNA. Transfer RNA (tRNA) molecules have numerous modified nucleosides. [0074] RNA virus and DNA virus detection test kits rely upon being able to identify a portion of the RNA sequence of an RNA virus or a portion of the DNA sequence of a DNA virus. RNA and DNA biopolymers found in animals, bacteria, archaea, mitochondria, chloroplasts, and viruses range in size from 21 nucleotides, such as small interfering RNA (siRNA), to large chromosomes in a single molecule that contain 247 million base pairs (human chromosome 1). Most living cells contain both DNA and RNA, with the exception of cells such as mature red blood cells. Viruses usually contain either DNA or RNA, but not both. If the virus is an RNA virus, then a reverse transcriptase enzyme process must be performed to prepare the corresponding DNA reverse transcript. After this, the DNA in the biological sample can be amplified (mass-produced) using a PCR (polymerase chain reaction) process. [0075] EXAMPLES [0076] The following Examples further illustrate the invention but are not to be construed as limiting its scope in any way. [0077] Example 1. Embodiment of the Present Invention Methods and Compositions for Testing for DNA Virus in a Person [0078] Some embodiments of the invention comprise an improved testing method for performing a DNA virus nucleic acid detection test on a biological sample, the improved testing method comprising the steps of: (a) obtaining a biological sample from a selected source of biological matter; (b) contacting the biological sample without delay with an amount of lysis buffer to form a mixture, wherein the lysis buffer comprises a protein denaturing agent in a fluid; (c) using the protein denaturing agent from the fluid lysis buffer in the mixture as a means to denature proteins immediately without refrigeration and keep the proteins inactivated in the mixture and prevented from degrading nucleic acids in the mixture; (d) using the protein denaturing agent from the fluid lysis buffer in the mixture to cause an immediate lysis of biological cells, microorganisms, bacteria, virus particles, and any other infectious organisms that may have come from the biological matter in the biological sample so as to sterilize the mixture immediately and safely; (e) storing optionally, the mixture of the biological sample and the fluid lysis buffer without refrigeration until needed for performing the DNA virus nucleic acid detection test; (f) transporting optionally, the mixture of the biological sample and the fluid lysis buffer without refrigeration to a location where the mixture of the biological sample and the fluid lysis buffer is needed for performing the DNA virus nucleic acid detection test; (g) using a purification means for obtaining purified nucleic acids from the mixture of the biological sample and the lysis buffer; (h) performing a PCR process using the purified nucleic acids with a probe and selected primers as a means for performing the DNA virus nucleic acid detection test on the biological sample. [0079] Example 2. An embodiment of the present invention methods and compositions for testing for RNA virus in a person [0080] Some embodiments of the invention comprise an improved testing method for performing an RNA virus nucleic acid detection test on a biological sample, the improved testing method comprising the steps of: (a) obtaining a biological sample from a selected source of biological matter; (b) contacting the biological sample without delay with an amount of lysis buffer to form a mixture, wherein the lysis buffer comprises a protein denaturing agent in a fluid; (c) using the protein denaturing agent from the fluid lysis buffer in the mixture as a means without refrigeration to denature proteins immediately and keep the proteins inactivated in the mixture and prevented from degrading nucleic acids in the mixture; (d) using the protein denaturing agent from the fluid lysis buffer in the mixture to cause an immediate lysis of biological cells, microorganisms, bacteria, virus particles, and any other infectious organisms that may have come from the biological matter in the biological sample so as to sterilize the mixture immediately and safely; (e) storing optionally, the mixture of the biological sample and the fluid lysis buffer without refrigeration until needed for performing the RNA virus nucleic acid detection test; (f) transporting optionally, the mixture of the biological sample and the fluid lysis buffer without refrigeration to a location where the mixture of the biological sample and the fluid lysis buffer is needed for performing the RNA virus nucleic acid detection test; (g) using a purification means for obtaining a purified nucleic acids sample from the mixture of the biological sample and the lysis buffer; (h) using a reverse transcription process to convert any ribonucleic acids (RNA) present in the purified nucleic acids sample into deoxyribonucleic acids (DNA) in the purified nucleic acids sample; (h) performing a PCR process using the purified nucleic acids with a probe and selected DNA primers to identify the DNA reverse transcribed from the RNA as a means for performing the RNA virus nucleic acid detection test on the biological sample. [0081] Example 3. Embodiment of the present invention test methods and compositions for testing for nonhuman DNA in a person [0082] Some embodiments of the invention comprise an improved testing method for performing a non-human DNA detection test on a biological sample, the improved testing method comprising the steps of: (a) obtaining a biological sample from a selected source of biological matter; (b) contacting the biological sample without delay with an amount of a lysis buffer to form a mixture, wherein the lysis buffer comprises a protein denaturing agent in a fluid; (c) using the protein denaturing agent from the fluid lysis buffer in the mixture as a means without refrigeration to denature proteins immediately and keep the proteins inactivated in the mixture and prevented from degrading nucleic acids in the mixture; (d) using the protein denaturing agent from the fluid lysis buffer in the mixture to cause an immediate lysis of biological cells, microorganisms, bacteria, virus particles, and any other infectious organisms that may have come from the biological matter in the biological sample so as to sterilize the mixture immediately and safely; (e) storing optionally, the mixture of the biological sample and the fluid lysis buffer without refrigeration until needed for performing the non-human DNA detection test; (f) transporting optionally, the mixture of the biological sample and the fluid lysis buffer without refrigeration to a location where the mixture of the biological sample and the fluid lysis buffer is needed for performing the nonhuman DNA detection test; (g) using a purification means for obtaining purified nucleic acids from the mixture of the biological sample and the lysis buffer; (h) performing a PCR process using the purified nucleic acids with a probe and selected primers as a means for performing the nonhuman DNA detection test on the biological sample. Example 4. Another embodiment of the present invention provides test methods and compositions for testing for nonhuman RNA in a person. Some embodiments of the invention comprise an improved testing method for performing a nonhuman RNA detection test on a biological sample, the improved testing method comprising the steps of: (a) obtaining a biological sample from a selected source of biological matter; (b) contacting the biological sample without delay with an amount of a lysis buffer to form a mixture, wherein the lysis buffer comprises a protein denaturing agent in a fluid; (c) using the protein denaturing agent from the fluid lysis buffer in the mixture as a means without refrigeration to denature proteins immediately and keep the proteins inactivated in the mixture and prevented from degrading nucleic acids in the mixture; (d) using the protein denaturing agent from the fluid lysis buffer in the mixture also to cause an immediate lysis of biological cells, microorganisms, bacteria, virus particles, and any other infectious organisms that may have come from the biological matter in the biological sample so as to sterilize the mixture immediately and safely; (e) storing optionally, the mixture of the biological sample and the fluid lysis buffer without refrigeration until needed for performing the nonhuman RNA detection test; (f) transporting optionally, the mixture of the biological sample and the fluid lysis buffer without refrigeration to a location where the mixture of the biological sample and the fluid lysis buffer is needed for performing the nonhuman RNA virus nucleic acid detection test; (g) using a purification means for obtaining a purified nucleic acids sample from the mixture of the biological sample and the lysis buffer; (h) using a reverse transcription process to convert any nonhuman ribonucleic acids (RNA) present in the purified nucleic acids sample into nonhuman deoxyribonucleic acids (DNA) in the purified nucleic acids sample; (h) performing a PCR process using the purified nucleic acids with a probe and selected nonhuman DNA primers to identify the nonhuman DNA reverse transcribed from the nonhuman RNA as a means for performing the nonhuman RNA nucleic acid detection test on the biological sample. [0083] Some embodiments of the present invention are designed to provide high- sensitivity true positive results, and to not provide false negative results. Some embodiments of the invention comprise buffer compositions useful as a safe viral lysis sample collection and testing buffer. After safely lysing the nucleic acids, then the identity of the nucleic acids can be tested using various nucleic acid tests using either known or novel chemical, biochemical and genetic biotechnology procedures which may include a selected PCR procedure, a colorimetric process, and/or a nucleic acid sequencing process with reference nucleic acids sequence(s) of a known genus, species, or mutant species of a life form or a virus. This is particularly useful for detecting the presence of a virus or viral infection in a person, in an animal, or a virus- contaminated location such as a surface, a fluid, or gas including respiratory or oral exhalations. [0084] FIG.1 depicts some initial steps of one embodiment of the present invention which is one method of adding a lysis buffer 110 as a spray 108 or as a buffer mist 108 onto a dry nucleic acids sample 109 on a surface 107 as a means for protecting nucleic acids to be tested 103 from various enzymes of nucleic acids (see Background for examples of the enzymes) as well as lysing viral capsules, bacterial walls, cell membranes or other membranes which may be confining the nucleic acids to be tested 103 in the sample 102, 106, or 109. For example, one method of delivering a spray formulation 108 of the lysis buffer 110 as a means for protecting nucleic acids to be tested 103 from various enzymes of nucleic acids may comprise the steps of: (a) depositing a wet sample specimen 102 with nucleic acids to be tested 103 upon a surface 101; (b) allowing optionally, the wet sample droplet 102 to situate as a wet sample 106 of the nucleic acids to be tested 103 on a surface 104 for a period of drying (wherein surface 104 which may be surface 101); and then (c) adding a lysis buffer 110 which comprises a formulation 108 by spraying from a container nozzle tip 111 upon a dried, semi-dry, or wet nucleic acids sample 109 which is situated on a surface 107 (which may have been the same surface as surface 101, or surface 104). The source of the wet nucleic acids sample specimen 102 may be a biological sample from any location or body biological material of a person or an animal. [0085] Optionally, as depicted in FIG.1, one or more added nucleic acid in the physical form of an oligonucleotide 105 may be added to the original nucleic acids sample to serve as an internal control, for relative quantification of copy numbers between the nucleic acid to be tested and the added nucleic acid(s), for quality control and tracking, for calibration of false negative or false positive sensitivity and probability, as a carrier nucleic acid, carrier RNA, carrier DNA, and any combination thereof, or for other purposes. [0086] Generally, for the present invention embodiments, a biological sample which contains nucleic acids to be tested 103 may come from a living or dead human, animal, or be an in vitro biological sample, or might be from an inanimate location. For example, the location of the biological sample containing nucleic acids to be tested 103 may be selected from the group consisting of a skin location, a hair, a wound, a sore, a blister, a scab, a finger nails scraping sample, a sweat sample, an eye, an inside cheek surface, a tongue surface, inside a nostril, throat, from another oral cavity location, a urine sample, a fecal matter sample, a blood sample, a blood plasma sample, a lymph sample, a cerebrospinal fluid sample, a sputum sample, a saliva sample, a vomit sample, a respiratory tract fluid or surfactant liquid sample, a human sample, an animal sample, a bone marrow sample, an internal organ, a body secretion, a tissue biopsy, a cell sample, a corpse, and any combination thereof. Nucleic acids to be tested 103 sampled from an inanimate object such as dry surface may need to be rubbed with an object that has been pre-wetted by the lysis buffer 110 or another lysis medium of the present invention. [0087] FIG.1 is an example procedure wherein method of biological sample mixture preparation uses a flat surface to which first added is biological matter to be lysed by spraying a lysis buffer on the biological matter to form biological sample mixture. [0088] Briefly stated, above FIG.1A depicts an example procedure wherein a wet sample specimen 102 comprising a nucleic acid to be tested 103 is provided, adding the wet sample specimen 102 comprising a nucleic acid to be tested 103 to a surface 101 and allowing it to dry or remain wet on a paper surface 104 as shown in FIG.1B, and in step (C) then spraying the components depicted in FIG.1B with a lysis buffer mist 108. Optionally the lysis buffer 108 may further comprise an added nucleic acid 105. As an example, the added nucleic acid 105 may comprise an internal control, a carrier nucleic acid, carrier RNA, carrier DNA, and any combination thereof. Optionally there may be pooling of samples. Surfaces 101, 104, and 107 may be same or different or/and may be flat or curved in any conceivable manner. The lysis buffer denatures proteins so as to inactivate ribonucleases / RNases, DNases and other related nucleic acid degrading enzymes likely present in the wet sample specimen 102. [0089] Generally, for the present invention embodiments, it is conceived that a biological sample which contains nucleic acid to be tested 103 may come from a living or dead human, animal, or be an in vitro biological sample, or might be from an inanimate location. For example, the location of the biological sample containing nucleic acid to be tested 103 may be selected from the group consisting of a skin location, a hair, a wound, a sore, a blister, a scab, a finger nails scraping sample, a sweat sample, an eye, an inside cheek surface, a tongue surface, inside a nostril, throat, from another oral cavity location, a urine sample, a fecal matter sample, a blood sample, a blood plasma sample, a lymph sample, a cerebrospinal fluid sample, a sputum sample, a saliva sample, a vomit sample, a respiratory tract fluid or surfactant liquid sample, a human sample, an animal sample, a bone marrow, an internal organ a body secretion, a tissue biopsy, a cell sample, a corpse, and any combination thereof. [0090] The protein concentration in saliva samples is 0.5-1.5 mg/ml. Assuming there is less than 1 ml saliva on the collection swab, then the protein amount is below 1 mg, and therefore the protein concentration in the lysis buffer can be below 1 mg protein/ml biological sample mixture (see FIG.2 description). [0091] For present invention embodiments, for example as depicted in FIG.1, the surface 101, surface 104, and surface 107 may be the same or different surfaces, and may be selected from the group consisting of a dry surface, a solvent-wetted surface, a surface wetted lysis buffer 110, a surface wetted with a large aqueous concentration of known-sequence polynucleotides as a polynucleotides or nucleic acid chum, a paper surface, a wax paper surface, a plastic surface, a test tube surface, a filter membrane surface, an Amicon filter surface, an Amicon filter surface for retaining polynucleotides with a length of greater than 100 nucleic acids bases on the surface of the Amicon filter surface, a 3K Amicon filter surface, a 10K Amicon filter surface, a 50K Amicon filter surface, a Millipore filter surface, an Millipore filter surface for retaining polynucleotides with greater than 100 nucleic acids base pairs on the surface of the Millipore filter surface, a glass surface, a Teflon surface, a silica surface, a diatomaceous earth surface, a chalk surface, a metal surface, a magnetic bead surface, a silica-coated magnetic bead surface, a surface with carboxylic acid groups, a surface with hydroxyl groups, a surface reversibly binding nucleic acids, a surface which binds nucleic acids and from which the nucleic acids may be subsequently eluted as needed, a test tube surface, a sample container or collection tube surface, a pipette tip surface, a syringe needle, a cotton swab fiber surface, a synthetic plastic swab surface, an agar gel surface, a gelatin surface, a fluid surface, an ice surface, a refrigerated surface, a glass slide surface, a microfluidics tube or chamber, an Oxford Nanopore nucleic acids sequencer chamber surface or port, an Oxford Nanopore nucleic acids sequencer device surface, a nucleic acids sequencer surface, and any combination thereof. [0092] The term ‘chum’ refers to carrier nucleic acids, such as carrier RNA or carrier DNA, which are added to the sample or reaction vessel to stabilize the actual biological sample or nucleic acid of interest. For example, the addition of carrier RNA is useful to reduce or prevent degradation of the RNA that is to be tested for. As an example, residual enzymes acting on nucleic acids, such as RNases, which may be present in the collected biological sample (or other mechanisms that result in the general degradation, adsorption or inactivation of nucleic acids, DNA or RNA) will be engaged by the chum. Chum is advantageously added at high concentrations for the purpose of protecting the nucleic acid, DNA or RNA that is to be tested for from degradation, adsorption or inactivation. Examples of concentrations of chum, such as added carrier RNA, are 10 pM – 1 nM, 1 nM – 100 nM, 100 nM – 10 µM, 10 µM – 1 mM and any combination thereof. As an example, the added chum or carrier RNA can alternatively be provided in the reagent buffer or in any other reaction component that is part of the testing procedure, such as in a tube or vessel that is pre-coated or pre- equipped with carrier RNA, positive or negative controls, or other blocking agents that stabilize RNA, DNA or nucleic acids from degradation, adsorption or inactivation. [0093] For some embodiments of the invention, nucleic acid to be tested 103 containing samples may be handled as depicted in FIG.1 and air-dried on a surface 101, a surface 104, or a surface 107 before or after spraying the immobilized sample specimen 106 with the lysis buffer mist 108. Optionally, for some embodiments of invention, the nucleic acid to be tested 103 sample may be dried by using a drying step comprising spray-drying, freeze-drying, using a desiccant or drying in a vacuum bell jar under vacuum. In addition, a refrigerating step may be useful for preserving some nucleic acids samples, the refrigerating step comprising storing the nucleic acid samples in a conventional refrigerator/freezer, storing them on ice or dry ice in a cooler, or storing in a subzero refrigerator or storing in a container cooled by liquid nitrogen. [0094] FIG.2 depicts another example procedure embodiment of the present invention wherein the method of biological sample mixture preparation utilizes a collection tube in which first added is lysis buffer and then added is biological matter to be lysed to form the biological sample mixture. [0095] In step (A) of FIG.2, a lysis buffer 208 optionally containing an added nucleic acid 205 is added to a biological sample collection tube 201. In step (B) of FIG.2, a nucleic acid to be tested 203 in biological matter such as fluid of a sample specimen 202 is added to collection tube 201 for immediately creating a biological sample mixture depicted in step (C) of FIG.2 which comprises the lysis buffer 208 and sample specimen 202, optionally further comprising an added nucleic acid 205, all of which are added to the sample collection tube 201. As an example, an added nucleic acid may comprise an internal control, a quality control, a negative control, a carrier nucleic acid, carrier RNA, carrier DNA, and any combination thereof. The biological sample mixture depicted in step (C) preferably is created essentially at the same time as the sample specimen 202 is collected from a person, using for example a collection swab. Present invention embodiments of the biological sample mixture may be of a great advantage in settings and countries that do not have refrigeration. [0096] Collection tubes 201 in steps (A), (B), and (C) preferably are the same collection tube 201. This fluid mixture of the lysis buffer 208 and specimen 202, and optionally with an added nucleic acid 205, is a novel composition embodiment of the present invention and a method comprising: creating immediately and efficiently a surprisingly stable biological sample not needing refrigeration. This fluid mixture is a nonobvious composition and method preparation step in view of the worldwide practices taught in 2020 by all health care emergency doctors, labs, CDC, and WHO officials and doctors whose edicts and standard operating procedures (SOP) for biological sample preparation for dealing with SARS-CoV-2 testing are quite different. Furthermore, inventor(s) have invented a biological sample mixture which usefully does not need refrigeration (refrigeration free method) as a means for effectively protecting any nucleic acids initially present in the biological sample by causing an inhibition of the degradation of the nucleic acids until the biological sample mixture can be tested for a pathogen such as SARS-CoV-2 virus. The present invention protects nucleic acids immediately during instant biological sample preparation as a part of the invention’s specimen collection process and the biological sample mixture embodiment of the present invention can be immediately prepared (created) anywhere rapidly and efficiently and safely as the biological matter is being collected from the person. FIG. 2 depicts a method for forming a composition which is a mixture which causes an inhibition of both possible rapid degradation and possible slow degradation of the nucleic acids in the biological sample from a biological matter of a person. This inhibition is caused by including a protein denaturing agent in the lysis buffer which acts immediately in the fluid mixture depicted in rightmost collection tube 201 depicted in FIG.2. Such embodiments of the present invention are nonobvious and novel biological sample mixture compositions and sample preparation methods because the currently available testing procedures do not use a lysis buffer and require refrigeration before and during shipping, transporting, and storing the prior art biological samples prepared until there is a time to perform a pathogen detection test using a PCR method which in the prior art depends upon PCR equipment. [0097] Furthermore, currently available methods involve unneeded dilution of the nucleic acids concentration in the collected biological specimen by an estimated between 1.25- fold to 5-fold. This causes prior art PCR tests more likely to report a false negative test result. Over dilution of the biological sample lowers test sensitivity and accuracy particularly if the person’s biological specimen is problematic in having low virus titer to start with. FIG.2 depicts a method for forming a composition which is a mixture which causes an inhibition of both possible rapid degradation and possible slow degradation of the nucleic acids in the biological sample from a biological matter of a person. This inhibition is caused by the lysis buffer 208 which comprises a fluid with a protein denaturing agent. The sample specimen mixture is a fluid in a collection tube 201 which would be in rightmost collection tube 201 depicted in FIG.2. In steps (B) and (C) of FIG.2, the lysis buffer 208 is capable of lysing cell walls of cells in the biological matter of sample specimen 202. [0098] Optionally there may be pooling of samples. Pooling of samples from a small group of people is an efficient way to find out if anyone in a larger group of people has been infected with the virus without using individual testing. If the test with pooled samples is found positive for the virus, then the testing would be repeated to find the individual(s) who is positive for the SARS-CoV-2 virus. [0099] FIG.3 shows an example procedure wherein the method of biological sample mixture preparation uses a flat surface to which first added is a lysis buffer and then added is biological matter to be lysed so as to form a biological sample mixture [00100] Depicted in FIGS.3A-3C is an example procedure wherein method of biological sample mixture preparation is a step of using a flat surface which is the same surface numbered 301, 304 and 307. Compared to FIG.1 method, FIG.3 step (A) is a step for adding the lysis buffer 308 optionally containing an added nucleic acid 305 to a flat surface 301 which is capable of absorbing or drying the lysis buffer 308. In FIG.3, step (B) is a step for adding a sample specimen 302 containing some nucleic acids to be tested 303 to surface 304 which already contains an amount of lysis buffer 308 which is labeled in step (B) as lysis buffer 309, applied or immobilized to flat surface 301. In FIG.3, step (C) depicted is the biological sample mixture composition which will have immediately denatured proteins and enzymes in the biological sample mixture as well as have lysed cell walls and organelle walls inside cells so as to permit a complete inactivation of ribonucleases, deoxyribonucleases, and other enzymes which if not inactivated by denaturing them, could rapidly degrade the nucleic acids from sample specimen 302. [00101] In one variation of the invention, the lysis buffer 308 is applied to personal protective equipment (PPE), such as surgical masks, air filtration devices or air filters, shields, suits, gloves, shoes, gowns, either worn by a person as part of PPE or installed in instruments or facilities, such that SARS-CoV-2 containing aerosols or particles are inactivated upon contact with the PPE. Protective equipment as defined here includes PPE as well as systems that protect more than one individual. Examples include systems for heating, ventilation, air conditioning / HVAC, air filtration, sterilization, recirculation of breathable air for use either inside or outside. These may for example be located in masks, suits, buildings, hospitals, offices, schools, sports and entertainment venues, shopping centers, stores, shops, markets, transportation systems, vehicles, cars, buses, trains, airplanes, sheets, room dividers, walls, floors, elevators, escalators. [00102] Optionally the lysis buffer may comprise reagents that undergo a visible color change after a swab, sputum or another biological sample containing the virus or the nucleic acid to be tested for is detected. Such a color change is useful to indicate a "SAFE" time after which the biological sample is inactivated and no longer infectious after being brought in contact with the lysis buffer or the dried/lyophilized components of the lysis buffer. As an example, the color change reaction may occur in the collection tube after the biological samples is added, on PPE, such as on a protective mask worn by a person, when she comes in contact with the biological sample, the virus or the nucleic acid to be tested for, or on equipment or instrumentation that may come in contact with the biological sample, the virus or the nucleic acid to be tested. [00103] FIG.4: Procedure wherein wet nucleic acids sample is added to a porous surface which may be a filter surface of any kind capable of retaining nucleic acids [00104] FIG.4, step (A) depicts an example procedure step for adding nucleic acid to be tested 403 sample specimen 402 to a porous surface 401 which may be a filter surface of any kind capable of retaining nucleic acids to some extent. A sample specimen 402 optionally is allowed to dry or is allowed remain wet on a filter surface 404as shown in step (B) and then in step (C) added is a lysis buffer 408 optionally containing an added nucleic acid405 with the immobilized biological sample mixture 406_depicted in step (C). Filter surfaces 401, 404, and 407 may be same or different or/and may be flat or curved in any conceivable manner. The lysis buffer 408 is for denaturing proteins in the biological sample mixture 40_ so as to inactivate ribonucleases, deoxyribonucleases and other related nucleic acid degrading enzymes likely present in the wet nucleic acids to be tested 403. Optionally there may be a pooling of sample specimens 402 so that more people may be tested at the same time. If a positive test result is found in pooled samples tested at the same time, then individual samples can be tested to identify the indivual with a SARS-CoV-2 infection. One definition of a biological sample mixture for the present invention is a biological sample mixture 40_ comprising a lysis buffer 408 mixed with a sample specimen 402 which comprises nucleic acids and other biological matter, and which may further comprise an added nucleic acid 405. [00105] FIG.5A: Procedure wherein a collection tube has contents which are a biological sample comprising a lysis buffer fluid with denatured proteins, lysed cell proteins, lipids, and nucleic acids, and optionally further comprising an added nucleic acid. [00106] FIG.5A depicts an example procedure step for placing into a sample collection tube 501 contents which comprise a lysis buffer fluid with sample specimen 502, comprising denatured proteins 510 and cellular debris, lysed proteins and lipids 510, and with nucleic acids to be tested 503, and optionally further comprising added nucleic acids 505. FIG.5 B depicts performing a non-magnetic isolation of the nucleic acid to be tested 503 by running a spin column 507 having a spin column material 509 in a collection tube 508. Added nucleic acids 505 may be retained along with nucleic acids to be tested 503 in the spin column material 509 or washed and discarded into the flow-through 506 materials which include denatured proteins 510 and cellular debris, lysed proteins and lipids 510, FIG.5C depicts the nucleic acids to be tested 503 and optional added nucleic acid 505 in nuclease-free water 511. [00107] FIG.5D depicts an example spin column procedure for purifying RNA and DNA from collection buffer mixture using ethanol for extracting the RNA and DNA and washing the nucleic acids free of the lysis buffer and denatured proteins and cellular debris. [00108] Another spin column procedure for purifying nucleic acids comprises the steps of: 1) adding a volume of absolute ethanol (100%) equivalent to 0.8x to 2x of the volume of the biological sample mixture (comprising the biological sample and lysis buffer and optionally an added nucleic acid) to the sample collection tube and mixing the sample of nucleic acids in the collection tube by vortexing; [00109] 2) transferring the mixture into a spin column in a collection tube 508 and centrifuging at >10,000 x g for one minute, then discarding the collection tube including the flow-through fluid; [00110] 3) transferring the spin column containing the spun down sample to a new collection tube and adding a volume between 400-700 µl ethanol (80%) to the spin column and centrifuging at 10,000 x g for one minute, then discarding the flow- through fluid; [00111] 4) adding a volume between 400-700 µl ethanol (80%) to the spin column and centrifuging at >10,000 x g for one minute, then discarding the flow-through fluid, wherein the centrifuging is continued at >10,000 x g for three minutes for drying the membrane; and [00112] 5) transferring the spin column to a clean micro-centrifuge tube and adding a volume between 10-100 µl of nuclease-free water into the spin column with then a centrifuging at >10,000 x g for one minute, wherein the eluted RNA/DNA is useable immediately for performing molecular based applications or storing at ‑80 ºC for using in the future. Alternatively the spin column procedure for purifying nucleic acids, instead of using ethanol, may use a hydroxylated organic chemical selected from the group consisting of isopropanol, methanol, n-propanol, a butyl alcohol, a pentyl alcohol, a hexyl alcohol, ethylene glycol, propylene glycol, glycerol, and any combination thereof. Optionally an alternative type of alcohol, polyalcohol or dehydrating agent can be used, such as toluene, nitromethane, ether and any combination thereof. [00113] FIG.6: Procedure for a magnetic bead-based purification of nucleic acids RNA and DNA from oral swab collected biological matter for SARS-CoV-2 virus [00114] FIG.6 depicts an example of a procedure for using a magnetic bead-based methods for purifying nucleic acids (RNA and DNA) from oral swabs collection of a biological matter for testing for SARS-CoV-2 virus particles (virions). One example of magnetic particles for the separation and purification of nucleic acids are described in: Magnetic particles for the separation and purification of nucleic acids, Berensmeier S, Applied Microbiology and Biotechnology volume 73, pages 495–504 (2006). [00115] Before FIG.6 step (C) but not depicted is a present invention example embodiment method step comprising first creating a biological sample by situating some biological material (not depicted) in form of a sample specimen collected by a collection means such as for example using a specimen swab (not depicted) into sample collection tube 601 and then adding an amount 100-1000 µl of a lysis buffer so that the sample collection tube 601 contains nucleic acids to be tested 603, denatured enzymes such as ribonucleases and deoxyribonucleases 610, cell debris, other denatured proteins 610, and lysed cells 610. The specimen swab (not depicted in FIG. 6 is swirled vigorously for 5-15 seconds, then as much liquid as possible is expunged from the specimen swab by pressing it to the wall of the sample collection tube 601. This collection of the specimen fluid from the specimen swab is not depicted in FIG. 6. The specimen swab is then discarded using a known safe disposal method for infectious pathogens. [00116] Depicted in FIG.6A is a step of adding reagent 604 containing magnetic beads 612 and optionally the added nucleic acid 605 to the collection tube 601 containing the sample specimen 602, which contains the nucleic acid to be tested 603 as well as cellular debris and other unwanted components 610. The mixture in collection tube 601 is mixed by pipetting it up and down, during which time the magnetic beads 612 bind to the nucleic acid to be tested 603 and optionally to the added nucleic acid 605 or control nucleic acids if present. [00117] Depicted in FIG.6B is a transferring of the collection tube 601 onto a magnetic rack 613 where the contents in the collection tube 601 are magnetized for 5 min or until the solution inside collection tube 601 appears clear. The magnetic rack 613 functions to pull the complex 614 comprising the magnetic beads 612, nucleic acid to be tested 603 and optionally the added nucleic acid 605 or control nucleic acids onto the inside wall of collection tube 601. Supernatant from collection tube 601 is removed (arrow 615) and discarded without disturbing the complex 614 comprising the bead pellet with nucleic acid to be tested 603 and optionally the added nucleic acid 605. Then (arrow 616) about 100 – 1000 µl (microliters) ethanol 617 are added to each collection tube 601. Do not mix. Then the supernatant containing ethanol is removed from collection tube 601 (see arrow 615) and discarded without disturbing the complex 614. Repeat this procedure. Then remove the collection tube 601 from the magnetic rack 613 and air dry the pellet complex 614 for 5-10 min. [00118] Depicted in FIG.6C is the step of adding (arrow 619) of a volume of between about 10 µl to about 100 µl of a nuclease-free water 618 to each collection tube 601 and then there is a pipetting of the sample up and down until the complex 614 is dissolved. Incubate the collection tube 601 for 5 min. Transfer the collection tube 601 containing complex 614 onto the magnetic rack 613 and incubate the sample for 2-10 min. or until the supernatant solution appears clear. [00119] Depicted in FIG.6D is the step of aspirating (see arrow 620) the supernatant 622 comprising the eluted nucleic acids to be tested 603 optionally the added nucleic acid 605 to a new collection tube 621. The eluted nucleic acids to be tested 603 and optionally the added nucleic acid 605 can be used immediately for molecular based applications or stored at -80ºC for future use. [00120] FIG.7: Purification of nucleic acids in a mixture of collected biological matter containing a lysis buffer through use of a centrifugal filter membrane [00121] Depicted in FIGS.7A-7D are method steps for purifying nucleic acids in a biological collection mixture through use of a 10-100 kDa centrifugal filter membrane. Nucleic acids that are larger than 50-500 base pairs that be retained. [00122] Not depicted in FIG.7A is the step of placing of the specimen swab containing in the swab fibers the source of the biological matter collected from a person preferably without a dilution into a collection tube with between about 100 µl to about 1000 µl of a lysis buffer embodiment of the present invention. There can be for example a swirling of the specimen swab containing the biological matter vigorously for 15 seconds, then an expunging as much liquid as possible from the specimen swab by pressing and rotating the fiber portion of the specimen swab against the wall of the collection tube. Discard the swab carefully as it may contain infectious pathogens. See previous figures and their description in the specification regarding biological matter collection locations in a person as well as the important inventive concept for avoiding dilution of biological matter and immediate uses of a lysis buffer. Note below FIG.9 depicts some examples of collection devices for collecting the biological matter specimen before adding a lysis buffer to the biological matter specimen. [00123] Depicted in FIG.7A is adding the sample specimen 702 containing cellular debris and other unwanted components as well as comprising the nucleic acid to be tested 703 and optionally the added nucleic acid 705 (with 703 and 705 together herein being collectively referred to as the nucleic acid batch 704) to a centrifugal filter membrane device 706 and insert it into an empty collection tube 701 with a cap 707. [00124] Depicted in FIG.7 step (B) is spinning the collection tube 701 containing the centrifugal filter membrane device 706 and the nucleic acid batch 704 in order to force the sample specimen 702 through the centrifugal filter membrane 709, thereby separating the sample specimen 702 into a retained fraction 708 comprising the nucleic acid batch 704, which is being retained by the centrifugal filter membrane 709, and the flow-through 710 containing cellular debris and other unwanted components, which passes the centrifugal filter membrane 709 and is collected at the bottom of tube 701. Discard the collection tube 701 and the flow-through 710. [00125] Depicted in FIG.7C is process including inverting the centrifugal filter membrane device 706, inserting it into a new empty collection tube 711 and spinning for recovering the retained fraction 708 comprising the nucleic acid batch 704 into collection tube 711. [00126] Depicted in FIG.7D is removing the centrifugal filter membrane device 706, for retaining collection tube 711 comprising the retained fraction 708 with the nucleic acid batch 704. [00127] FIG.8A depicts a reverse transcriptase PCR (RT-PCR) process: See: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7197457/ https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0215756 https://www.promega.com/resources/guides/nucleic-acid-analysis/pcr-amplification/ [00128] FIG.8B depicts a reverse transcriptase PCR (RT-PCR) process using viral RNA: [00129] FIG.8C depicts a reverse transcriptase PCR (RT-PCR) process. [00130] FIG.9 below depicts in accordance with some embodiments of the present invention, several common kinds of biological matter (sample specimen) collection devices. Depicted are: a device which is collection swab 901; a device 902 which is a collection swab 904 in a protective test tube 903 for protecting the collection swab 904; a device which is a test tube 907 with cork 906 to hold a fluidic biological matter specimen 908; and a device which is a syringe 909 to draw fluid samples from a person’s body, a liquid biological matter specimen 910, a biopsy sample 912 on a microscope slide or surface 911, or a biopsy sample 913. Biological matter from the collection device is preferably immediately (without delay) added to a lysis buffer to more effectively accomplish various objects of the present invention, including: (1) using the lysis buffer for inactivating any infectious pathogen in the biological matter sample specimen, and (2) using the lysis buffer for lysing cells and denaturing various proteins including enzymes and the like as a means for inhibiting degradation of the nucleic acids which may be present in the biological matter sample specimen. When these objects of the present invention are accomplished by forming a mixture which comprises the biological sample with the lysis buffer, then (3) the mixture being lysed by the lysis buffer need not be refrigerated (to inhibit enzymes such as ribonucleases and preserve the biological cells and the nucleic acids in the sample). Without the need for refrigeration the nucleic acids in the lysed biological matter sample can be transported, shipped, stored from any collection without the need for dry ice or ice. This for example frees up the collection sampling in remote third world locations, and even would allow self-sampling by a person needing to know if they have a SARS- CoV-2 virus infection. The mixture also represents a useful means for performing a minimal dilution of the nucleic acids in the biological matter specimen step as a means for allowing subsequent steps of running the nucleic acids detection testing method to be performed with a larger amount of the nucleic acids of the biological matter specimen. Reducing the dilution of the tested nucleic acids is useful for increasing the number of for example viral particles attempted to be detected in the test and as a result lowers risks of false negative test results. [00131] FIG.10 depicts a sequencing procedure to identify nucleic acid mutations for samples giving a positive nucleic acid test result. Sequencing procedures are described in the references listed below: https://www.promega.com/products/sequencing/?_ga=2.257415947.1854990525.1595 326675-2081187507.1595326675 https://www.nature.com/articles/s41598-019-41830-w https://www.illumina.com/content/dam/illumina- marketing/documents/products/illumina_sequencing_introduction.pdf https://www.researchgate.net/publication/282061980_Next_generation_sequencing_applicati ons_for_breast_cancer_research/figures?lo=1 https://www.google.com/imgres?imgurl=https%3A%2F%2Fwww.researchgate.net%2Fprofil e%2FAxel_Colling%2Fpublication%2F303499043%2Ffigure%2Ffig1%2FAS%3A56797569 9009540%401512427367904%2FEssential-steps-in-the-workflow-of-an-NGS-based-assay- from-sample-to-result-The- figure_Q320.jpg&imgrefurl=https%3A%2F%2Fwww.researchgate.net%2Fpublication%2F3 03499043_Next- generation_sequencing_workflows_in_veterinary_infection_biology_Towards_validation_an d_quality_assurance&tbnid=lAOJokK_9iCoPM&vet=12ahUKEwiXyqXOkN7qAhWIBt8K HRxeD9QQMygHegUIARCNAQ..i&docid=nholVWTSV0qJ1M&w=320&h=320&q=next% 20generation%20sequencing%20workflow%20multiplexed%20viral%20samples%20to%20d etermine%20if%20the%20biological%20sample%20had%20contained%20nucleic%20acids &ved=2ahUKEwiXyqXOkN7qAhWIBt8KHRxeD9QQMygHegUIARCNAQ [00132] Hot Start Polymerase - In the one step reverse-transcription PCR, the activity of the Polymerase during the reverse transcription at 50°C is blocked by an antibody or aptamers. After the reverse-transcription the mixture is heated up to 95°C to activate the Polymerase and inactivate the Reverse-Transcriptase. [00133] The heat stable Taq (‘AmpliTaq’) enzyme has a 5’-exonucleolytic function that does not displace the probe sitting in the middle but instead digests it, releasing the reporter dye in the process. Phi29 is a polymerase that is not heat stable (40C max) but which has a strand displacement function. Phi29 and related polymerases (such as the thermoresistant BST polymerase I) are used for isothermal amplification processes such as whole-genome amplification (WGA) or Loop-mediated isothermal amplification (LAMP). [00134] Bst DNA Polymerase is a thermoresistant polymerase primarily used for Loop-Mediated Isothermal Amplification (LAMP) that has been modified to eliminate 5´ →3´ exonuclease activity while retaining its 5´ → 3´ polymerase activity at elevated temperatures, around 65°C. It is suitable for applications requiring thermophilic strand displacement polymerase activity and gap-filling reactions and provides high product yields in demanding conditions. It is in particularly suitable for use with oligonucleotide primers that have not been protected from 3’-exonuclease digestion, such as by the addition of one or more phosphorothioate bonds during oligonucleotide synthesis, thereby making the synthesis of such oligonucleotides considerably cheaper. A phosphorothioate bond substitutes a sulfur atom for a non-bridging oxygen in the phosphate backbone of an oligonucleotide. This modification renders the inter- nucleotide linkage resistant to nuclease degradation. Phosphorothioate bonds can be introduced between the last 3-5 nucleotides at the 5'- or 3'-end of the oligonucleotide to inhibit exonuclease degradation. [00135] The table below. provides examples of useful primers and probes for practicing the present Invention.:
Figure imgf000037_0001
[00136] FIG.11 depicts an overview of the steps in Next Generation Sequencing workflow. [00137] FIG.12 is a schematic of steps in the workflow of a next generation sequencing based assay. [00138] Control Test Samples Needed for Nucleic Acids Testing: [00139] Various standard controls need to be run by the testing procedure, including testing known amounts of nucleic acids in an internal control test sample. One example for an internal control test is a specific sequence of nucleic acid, typically DNA or RNA, that is added to the testing procedure at a known concentration or copy number to ensure that the testing procedure works reliably and is able to detect both the positive control(s) and the sample at hand with the expected sensitivity. For example, it is critical to include appropriate positive controls in a quantitative PCR (qPCR) experiment to determine if false negatives are being detected in the experiment. Positive controls fall into two classes: [00140] 1. Exogenous internal controls refer to the use of external DNA or RNA carrying a target of interest. If these positive controls are assayed in separate wells/tubes from the experimental sample, they serve as a control to determine whether the reverse transcription and/or PCR reaction conditions are optimal or not. Additionally, exogenous DNA or RNA positive controls may be spiked into the experimental sample(s) and assayed in parallel or in a multiplex format with the target of interest. These control reactions assess whether the samples contain any components that inhibit reverse transcription and/or PCR. One example of an exogenous control in a sample of a patient being tested for SARS-CoV-2 is nucleic acid of a specific sequence that is added or spiked into the reaction in which the testing is performed. [00141] For example, an exogenous control can comprise one or more artificial sequences of nucleic acid, DNA or RNA that do not occur naturally in the biological samples being tested. It is advantageous to include several exogenous controls for reliability and redundancy. In order to obtain a more accurate estimate of the patient’s virus titer or copy number, it is further advantageous to include several exogenous controls at different concentrations or copy numbers, thereby allowing for a direct comparison of viral or pathogen load based on the titers or copy numbers of the various exogenous control or controls. In one example an exogenous control is provided at the following concentrations: 1 - 100fM, 100fM - 10pM, 10pM – 1nM, 1nM – 100nM, 100nM – 10µM, 10µM – 1mM, and any combination thereof. In another example an exogenous control is provided at the following copy numbers: 1, 2- 5, 5-10, 10-100, 100-1000, 1000-10E4 (10E4 = 10,000), 10E4 -10E5, 10E5-10E6, 10E6-10E7, 10E7-10E8, 10E8-10E9, 10E9-10E10, 10E10-10E11, 10E11-10E12, 10E12-10E13, 10E13-10E14, 10E14-10E15, 10E15-10E16, 10E16-10E17, 10E17- 10E18, and any combination thereof. [00142] An internal or negative control can be provided either in liquid or dried form. Several internal or negative controls can be provided in combination with either the same or at different concentrations or copy numbers. In one example the relative ratio between two or more internal or negative controls can be as follows: 1:1-1:2, 1:2-1:3, 1:3-1:4, 1:4-1:5, 1:5-1:6, 1:6-1:7, 1:7-1:8, 1:8-1:9, 1:9-1:10, 1:10-1:20, 1:20-1:30, 1:30-1:40, 1:40-1:50, 1:50-1:60, 1:60-1:70, 1:70-1:80, 1:80-1:90, 1:90-1:100, 1:100- 1:200, 1:200-1:500, 1:500-1:1000, 1:1000-1:2000, 1:2000-1:5000, 1:5000-1:10E4 (10E4 = 10,000), 1:10E4-1:10E5, 1:10E5-1:10E6, 1:10E6-1:10E7, 1:10E7-1:10E8, 1:10E8-1:10E9, and any combination thereof. [00143] 2. Endogenous internal controls refer to the use of a native target that is present in the experimental sample(s) of interest but is different from the target under study. These types of controls are often referred to as normalizers, and are typically used to correct for quantity and quality differences between samples. One example of an endogenous control in a sample of a patient being tested for SARS-CoV-2 is nucleic acid that is already present in the patient’s saliva, sputum, bodily fluid, tissue or from any other of the types and sources of biological samples described herein, but not originating from the actual SARS-CoV-2 virus. The use of an endogenous control can be useful to determine and quantify the efficiency of the biological sample collection, in other words as a measure of how effectively endogenous and viral material was acquired during the sample collection procedure. Doing so can provide a more accurate estimate of the patient’s virus titer or copy number through normalization based on the titer or copy number of the endogenous control locus or loci. In one example an endogenous control is selected from a suitable nucleic acid that is present at the following concentrations: 1 - 100fM, 100fM - 10pM, 10pM – 1nM, 1nM – 100nM, 100nM – 10µM, 10µM – 1mM and any combination thereof. [00144] Internal or negative controls can be included either in reagents or in reaction vessels, tubes or wells that are used to detect a specific sequence. As an example, reaction wells may include assays that detect artificial positive or negative controls spiked into each sample during a cDNA synthesis step, thereby ensuring that the reverse transcription step proceeded as needed. [00145] FIG 13. depicts some of the steps which may be used for some embodiments of the present invention for collecting cellular biological matter specimen in a collection tube, lysing cells in the cellular biological matter specimen in the collection tube, binding RNA from the collected cellular biological matter specimen to a column with a material which binds RNA, and washing and eluting the RNA from the RNA binding column into a tube for holding the RNA in a fluid. [00146] Virus Characteristics and Definitions Relating to Viruses [00147] Viruses display a wide diversity of shapes and sizes, called 'morphologies'. In general, viruses are much smaller than bacteria. Most viruses that have been studied have a diameter between 20 and 300 nanometers. Some filoviruses have a total length of up to 1400 nm; their diameters are only about 80 nm. Most viruses cannot be seen with an optical microscope, so scanning and transmission electron microscopes are used to visualize them. To increase the contrast between viruses and the background, electron-dense "stains" are used. These are solutions of salts of heavy metals, such as tungsten, that scatter the electrons from regions covered with the stain. When individual virus particles, called virions, are coated with stain (positive staining), fine detail is obscured. Negative staining overcomes this problem by staining the background only. (Virus, Wikipedia, 2020) [00148] A complete virus particle consists of nucleic acid surrounded by a protective coat of protein called a capsid. These are formed from identical protein subunits called capsomeres. Viruses can have a lipid "envelope" derived from the host cell membrane. The capsid is made from proteins encoded by the viral genome and its shape serves as the basis for morphological distinction. Virally-coded protein subunits will self- assemble to form a capsid, in general requiring the presence of the virus genome. Complex viruses code for proteins that assist in the construction of their capsid. Proteins associated with nucleic acid are known as nucleoproteins, and the association of viral capsid proteins with viral nucleic acid is called a nucleocapsid. The capsid and entire virus structure can be mechanically probed through atomic force microscopy. (Virus, Wikipedia, 2020). [00149] Helical Virus - These viruses are composed of a single type of capsomere stacked around a central axis to form a helical structure, which may have a central cavity, or tube. This arrangement results in rod-shaped or filamentous virions which can be short and highly rigid, or long and very flexible. The genetic material (typically single-stranded RNA, but ssDNA in some cases) is bound into the protein helix by interactions between the negatively charged nucleic acid and positive charges on the protein. Overall, the length of a helical capsid is related to the length of the nucleic acid contained within it, and the diameter is dependent on the size and arrangement of capsomeres. The well-studied tobacco mosaic virus is an example of a helical virus. (Virus, Wikipedia, 2020). [00150] Icosahedral Virus - Most animal viruses are icosahedral or near-spherical with chiral icosahedral symmetry. A regular icosahedron is the optimum way of forming a closed shell from identical sub-units. The minimum number of identical capsomeres required for each triangular face is 3, which gives 60 for the icosahedron. Many viruses, such as rotavirus, have more than 60 capsomers and appear spherical yet retain symmetry. To achieve this, the capsomeres at the apices are surrounded by five other capsomeres and are called pentons. Capsomeres on the triangular faces are surrounded by six others and are called hexons. Hexons are essentially flat and pentons, which form the 12 vertices, are curved. The same protein may act as the subunit of both the pentamers and hexamers or they may be composed of different proteins. (Virus, Wikipedia, 2020). [00151] Virus Envelope - Some species of virus envelop themselves in a modified form of one of the cell membranes, either the outer membrane surrounding an infected host cell or internal membranes such as nuclear membrane or endoplasmic reticulum, thus gaining an outer lipid bilayer known as a viral envelope. This membrane is studded with proteins coded for by the viral genome and host genome; the lipid membrane itself and any carbohydrates present originate entirely from the host. The influenza virus and HIV use this strategy. Most enveloped viruses are dependent on the envelope for their infectivity. (Virus, Wikipedia, 2020). [00152] Complex viruses possess a capsid that is neither purely helical nor purely icosahedral, and that may possess extra structures such as protein tails or a complex outer wall. Some bacteriophages, such as Enterobacteria phage T4, have a complex structure consisting of an icosahedral head bound to a helical tail, which may have a hexagonal base plate with protruding protein tail fibers. This tail structure acts like a molecular syringe, attaching to the bacterial host and then injecting the viral genome into the cell. [00153] The poxviruses are large, complex viruses that have an unusual morphology. The viral genome is associated with proteins within a central disc structure known as a nucleoid. The nucleoid is surrounded by a membrane and two lateral bodies of unknown function. The virus has an outer envelope with a thick layer of protein studded over its surface. The whole virion is slightly pleomorphic, ranging from ovoid to brick-shaped. [00154] Mimivirus is one of the largest characterized viruses, with a capsid diameter of 400 nm. Protein filaments measuring 100 nm project from the surface. The capsid appears hexagonal under an electron microscope, therefore the capsid is probably icosahedral. In 2011, researchers discovered the largest then known virus in samples of water collected from the ocean floor off the coast of Las Cruces, Chile. Provisionally named Megavirus chilensis, it can be seen with a basic optical microscope. In 2013, genus Pandoravirus was discovered in Chile and Australia, and has genomes about twice as large as Megavirus and Mimivirus. All giant viruses have dsDNA genomes and they are classified into several families: Mimiviridae, Pithoviridae, Pandoraviridae, Phycodnaviridae, and the Mollivirus genus. [00155] An enormous variety of genomic structures can be seen among viral species; as a group, they contain more structural genomic diversity than plants, animals, archaea, or bacteria. There are millions of different types of viruses, although fewer than 7,000 types have been described in detail. As of September 2015, the NCBI Virus genome database has more than 75,000 complete genome sequences, but there are doubtlessly many more to be discovered. (Virus, Wikipedia, 2020). [00156] A virus has either a DNA or an RNA genome and is called a DNA virus or an RNA virus, respectively. The vast majority of viruses have RNA genomes. Plant viruses tend to have single-stranded RNA genomes and bacteriophages tend to have double-stranded DNA genomes. (Virus, Wikipedia, 2020). [00157] Viral genomes are circular, as in the polyomaviruses, or linear, as in the adenoviruses. The type of nucleic acid is irrelevant to the shape of the genome. Among RNA viruses and certain DNA viruses, the genome is often divided up into separate parts, in which case it is called segmented. For RNA viruses, each segment often codes for only one protein and they are usually found together in one capsid. All segments are not required to be in the same virion for the virus to be infectious, as demonstrated by brome mosaic virus and several other plant viruses. (Virus, Wikipedia, 2020). [00158] A viral genome, irrespective of nucleic acid type, is almost always either single-stranded or double-stranded. Single-stranded genomes consist of an unpaired nucleic acid, analogous to one-half of a ladder split down the middle. Double-stranded genomes consist of two complementary paired nucleic acids, analogous to a ladder. The virus particles of some virus families, such as those belonging to the Hepadnaviridae, contain a genome that is partially double-stranded and partially single-stranded. (Virus, Wikipedia, 2020). [00159] For most viruses with RNA genomes and some with single-stranded DNA genomes, the single strands are said to be either positive-sense, called the 'plus-strand', or negative-sense, called the 'minus-strand', depending on if they are complementary to the viral mRNA. Positive-sense viral RNA is in the same sense as viral mRNA and thus at least a part of it can be immediately translated by the host cell. Negative-sense viral RNA is complementary to mRNA and thus must be converted to positive-sense RNA by an RNA-dependent RNA polymerase before translation. DNA nomenclature for viruses with single-sense genomic ssDNA is similar to RNA nomenclature, in that positive-strand viral ssDNA is identical in sequence to the viral mRNA and is thus a coding strand, while negative-strand viral ssDNA is complementary to the viral mRNA and is thus a template strand. Several types of ssDNA and ssRNA viruses have genomes that are ambisense in that transcription can occur off both strands in a double- stranded replicative intermediate. Examples include Gemini viruses, which are ssDNA plant viruses, and Arenaviruses, which are ssRNA viruses of animals (Virus, Wikipedia, 2020). [00160] Genome size varies greatly between species. The smallest—the ssDNA circoviruses, family Circoviridae—code for only two proteins and have a genome size of only two kilobases; the largest—the Pandora viruses—have genome sizes of around two megabases which code for about 2500 proteins. Virus genes rarely have introns and often are arranged in the genome so that they overlap. (Virus, Wikipedia, 2020). [00161] In general, RNA viruses have smaller genome sizes than DNA viruses because of a higher error-rate when replicating and have a maximum upper size limit. Beyond this, errors when replicating render the virus useless or uncompetitive. To compensate, RNA viruses often have segmented genomes—the genome is split into smaller molecules—thus reducing the chance that an error in a single-component genome will incapacitate the entire genome. In contrast, DNA viruses generally have larger genomes because of the high fidelity of their replication enzymes. Single-strand DNA viruses are an exception to this rule, as mutation rates for these genomes can approach the extreme of the ssRNA virus case. (Virus, Wikipedia, 2020). [00162] Genetic mutation - Antigenic shift, or re-assortment, can result in novel and highly pathogenic strains of human flu. Viruses undergo genetic change by several mechanisms. These include a process called antigenic drift where individual bases in the DNA or RNA mutate to other bases. Most of these point mutations are "silent"— they do not change the protein that the gene encodes—but others can confer evolutionary advantages such as resistance to antiviral drugs. Antigenic shift occurs when there is a major change in the genome of the virus. This can be a result of recombination or re-assortment. When this happens with influenza viruses, pandemics may result. RNA viruses often exist as quasi-species or swarms of viruses of the same species but with slightly different genome nucleoside sequences. Such quasi-species are a prime target for studying natural selection and the evolution of replicating RNA, such as from a virus (Fluoreszenzdetektion molekularer Evolution, Dapprich J, 1994, Cuvillier Verlag Göttingen, ISBN 3-89588-045-0, and: Virus, Wikipedia, 2020).Segmented genomes confer evolutionary advantages; different strains of a virus with a segmented genome can shuffle and combine genes and produce progeny viruses that have unique characteristics. This is called re-assortment or 'viral sex'. (Virus, Wikipedia, 2020). [00163] Genetic recombination is the process by which a strand of DNA is broken and then joined to the end of a different DNA molecule. This can occur when viruses infect cells simultaneously and studies of viral evolution have shown that recombination has been rampant in the species studied. Recombination is common to both RNA and DNA viruses. (Virus, Wikipedia, 2020). [00164] Viral populations do not grow through cell division, because they are acellular. Instead, they use the machinery and metabolism of a host cell to produce multiple copies of themselves, and they assemble in the cell. When infected, the host cell is forced to rapidly produce thousands of identical copies of the original virus. (Virus, Wikipedia, 2020). [00165] Basic virus life cycle comprises: (1) attachment, (2) penetration, (3) uncoating, (4) replication, (5) assembly, and (6) release. [00166] (1) Attachment is a specific binding between viral capsid proteins and specific receptors on the host cellular surface. This specificity determines the host range and type of host cell of a virus. For example, HIV infects a limited range of human leucocytes. This is because its surface protein, gp120, specifically interacts with the CD4 molecule—a chemokine receptor—which is most commonly found on the surface of CD4+ T-Cells. This mechanism has evolved to favor those viruses that infect only cells in which they are capable of replication. Attachment to the receptor can induce the viral envelope protein to undergo changes that result in the fusion of viral and cellular membranes, or changes of non-enveloped virus surface proteins that allow the virus to enter. (Virus, Wikipedia, 2020). [00167] (2) Penetration by plant virus of plant cells, fungal virus penetration of fungal cells, bacterial virus penetration of bacterial cell, and animal virus penetration of animal cells follows viral attachment to these cell types. Virions enter the host cell through receptor-mediated endocytosis or membrane fusion in a process known as viral entry. The infection of plant and fungal cells is different from that of animal cells. Plants have a rigid cell wall made of cellulose, and fungi one of chitin, so most viruses can get inside these cells only after trauma to the cell wall. Nearly all plant viruses can also move directly from cell to cell, in the form of single-stranded nucleoprotein complexes, through pores called plasmodesmata. Bacteria, like plants, have strong cell walls that a virus must breach to infect the cell. Given that bacterial cell walls are much thinner than plant cell walls due to their much smaller size, some viruses have evolved mechanisms that inject their genome into the bacterial cell across the cell wall, while the viral capsid remains outside. (Virus, Wikipedia, 2020). [00168] (3) Viral Uncoating – Uncoating is the process in which the viral capsid is removed: This may be by degradation by viral enzymes or host enzymes or by simple dissociation; the end-result is the releasing of the viral genomic nucleic acid. (Virus, Wikipedia, 2020). [00169] (4) Viral Replication - Replication of viruses primarily involves multiplication of the genome. Replication involves synthesis of viral mRNA from "early" genes (with exceptions for positive sense RNA viruses), viral protein synthesis, possible assembly of viral proteins, then viral genome replication mediated by early or regulatory protein expression. This may be followed, for complex viruses with larger genomes, by one or more further rounds of mRNA synthesis: "late" gene expression is, in general, of structural or virion proteins. (Virus, Wikipedia, 2020). [00170] (5) Assembly – Following the structure-mediated self-assembly of the virus particles, some modification of the proteins often occurs. In viruses such as HIV, this modification, also referred to as maturation, occurs after the virus has been released from the host cell. (Virus, Wikipedia, 2020). [00171] (6) Release – Viruses can be released from the host cell by lysis, a process that kills the cell by bursting its membrane and cell wall if present: this is a feature of many bacterial and some animal viruses. Some viruses undergo a lysogenic cycle where the viral genome is incorporated by genetic recombination into a specific place in the host's chromosome. The viral genome is then known as a "provirus" or, in the case of bacteriophages a "prophage". Whenever the host divides, the viral genome is also replicated. The viral genome is mostly silent within the host. At some point, the provirus or prophage may give rise to active virus, which may lyse the host cells. Enveloped viruses (e.g., HIV) typically are released from the host cell by budding. During this process, the virus acquires its envelope, which is a modified piece of the host's plasma or other internal membrane. (Virus, Wikipedia, 2020). [00172] Genome replication - The genetic material within virus particles, and the method by which the material is replicated, varies considerably between different types of viruses. [00173] DNA viruses - The genome replication of most DNA viruses takes place in the nucleus of the cell. If the cell has the appropriate receptor on its surface, these viruses enter the cell either by direct fusion with the cell membrane (e.g., herpesviruses) or— more usually—by receptor-mediated endocytosis. Most DNA viruses are entirely dependent on the host cell's DNA and RNA synthesizing machinery, and RNA processing machinery. Viruses with larger genomes may encode much of this machinery themselves. In eukaryotes the viral genome must cross the cell's nuclear membrane to access this machinery, while in bacteria it need only enter the cell. (Virus, Wikipedia, 2020). [00174] RNA viruses - Replication of RNA viruses usually occurs in the cytoplasm. RNA viruses can be placed into four different groups depending on their modes of replication. The polarity (whether or not it can be used directly by ribosomes to make proteins) of single-stranded RNA viruses largely determines the replicative mechanism; the other major criterion is whether the genetic material is single-stranded or double-stranded. All RNA viruses use their own RNA replicase enzymes to create copies of their genomes. (Virus, Wikipedia, 2020). [00175] Reverse transcribing viruses - Reverse transcribing viruses have ssRNA (Retroviridae, Metaviridae, Pseudoviridae) or dsDNA (Caulimoviridae, and Hepadnaviridae) in their particles. Reverse transcribing viruses with RNA genomes (retroviruses) use a DNA intermediate to replicate, whereas those with DNA genomes (pararetroviruses) use an RNA intermediate during genome replication. Both types use a reverse transcriptase, or RNA-dependent DNA polymerase enzyme, to carry out the nucleic acid conversion. Retroviruses integrate the DNA produced by reverse transcription into the host genome as a provirus as a part of the replication process; pararetroviruses do not, although integrated genome copies of especially plant pararetroviruses can give rise to infectious virus. They are susceptible to antiviral drugs that inhibit the reverse transcriptase enzyme, e.g. zidovudine and lamivudine. An example of the first type is HIV, which is a retrovirus. Examples of the second type are the Hepadnaviridae, which includes Hepatitis B virus. (Virus, Wikipedia, 2020). [00176] Cytopathic effects on the host cell - The range of structural and biochemical effects that viruses have on the host cell is extensive. These are called 'cytopathic effects'. Most virus infections eventually result in the death of the host cell. The causes of death include cell lysis, alterations to the cell's surface membrane, and apoptosis. Often cell death is caused by cessation of its normal activities because of suppression by virus-specific proteins, not all of which are components of the virus particle. The distinction between cytopathic and harmless is gradual. Some viruses, such as Epstein– Barr virus, can cause cells to proliferate without causing malignancy, while others, such as papillomaviruses, are established causes of cancer. (Virus, Wikipedia, 2020). [00177] Dormant and latent infections - Some viruses cause no apparent changes to the infected cell. Cells in which the virus is latent and inactive show few signs of infection and often function normally. This causes persistent infections and the virus is often dormant for many months or years. This is often the case with herpes viruses. (Virus, Wikipedia, 2020). [00178] Host range - Viruses are by far the most d biological entities on Earth, outnumbering all the others put together. They infect all types of cellular life including animals, plants, bacteria, and fungi. Different types of viruses can infect only a limited range of hosts and many are species-specific. Some, such as smallpox virus, can infect only one species, in this case humans, and are said to have a narrow host range. Other viruses, such as rabies virus, can infect different species of mammals and are said to have a broad range. The viruses that infect plants are harmless to animals, and most viruses that infect other animals are harmless to humans. The host range of some bacteriophages is limited to a single strain of bacteria, which can be used to trace the source of outbreaks of infections by a method called phage typing. The complete set of viruses in an organism or habitat is called the virome; for example, all human viruses constitute the human virome. (Virus, Wikipedia, 2020). [00179] The term nucleic acids shall mean sequences of nucleic acids containing a few nucleotides to thousands of nucleotides, wherein the nucleic acids further may comprise a single or multiple polymers in a sample and may comprise double stranded nucleic acids conformations or single strand nucleic acids conformations. [00180] Biological samples generally contain a large number of different enzymes which can rapidly degrade nucleic acids in various ways., Worldwide during the COVID-19 pandemic, available methods for detecting SARS-CoV-2 are problematic and do not provide the methods and compositions of the present invention in numerous ways, including biological sample collection guidelines for medical technicians, researchers, hospital staff, nurses, and doctors do not provide the lysis buffer compositions of the present invention. Specifically health authorities working for the CDC and WHO teach that SARS-CoV-2 sampling and testing of the sample should use conventional methods including: (1) a conventional method of collecting biological matter from a person and placing the biological matter immediately into several (1 to 3 ml) milliliters of a CDC VTM (Virus Transfer Medium) buffer or an aqueous phosphate buffered saline (PBS); and (2) then a conventional method of using the VTM or PBS buffer for a period of days to weeks to maintain a state of freshness in the biological samples. This conventional method unwittingly maintains a potential for virus particles, if present in the biological sample, to be capable of known viral activities such as inadvertent infection of workers handling the sample or transmission to other samples resulting in cross-contamination and reduced accuracy. Such uncontrolled viral activity is a potential direct health hazard for personnel collecting, storing, transporting and disposing of samples and presents substantial risks especially to those working with the samples in a laboratory setting and doing the actual testing. This conventional method also maintains the risk of unnecessarily allowing potentially rapid degradation of nonhuman nucleic acids (DNA and/or RNA) in the biological sample prior to testing for the presence of virus particles or nucleic acids to be tested. Thus, conventional treatment of biological samples prior to testing for the presence of non-human nucleic acid sequences increases the risk of false negative test results for the presence of non-human nucleic acids in a person. Thus, conventional methods of, for example, SARS-CoV-2 testing, can fail to provide an accurate result regarding the infectious status of the person who provided the biological matter for the biological sample as a means to determine whether they are infected with the SARS-CoV-2 virus or not. Prior art methods allow cells from the biological sample containing active nucleic acid hydrolyzing enzymes, such as RNases and DNases, to remain intact, creating the potential for inaccurate and unreliable test results. The present invention uses a lysis buffer to inactivate the nucleic acid hydrolyzing enzymes by quickly rendering and keeping these proteins in denatured inactive conformations, thereby increasing the accuracy of test results. [00181] Currently available methods teach refrigerating biological samples until testing for SARS-CoV-2 Virus. The CDC has indicated that during the shipping process, most molecular test swabs must be kept within a certain temperature range and must arrive at the lab within 72 hours to improve test accuracy.. More specifically, that the biological samples must be stored, shipped, or transported at a temperature near freezing (2-8 oC), or kept cooled below freezing temperatures until the biological sample is tested, which is sometimes days later. [00182] To prevent the rapid degradation of nucleic acids in the biological sample, some embodiments of the present invention practice a method including obtaining a biological sample from a selected source of biological matter; contacting the biological sample without delay with an amount of a lysis buffer(s) to form a mixture(s), wherein the lysis buffer comprises a protein denaturing agent in a fluid, and using the protein denaturing agent(s) from the fluid lysis buffer in the mixture as a means for storing the mixture for a time period of storage or transporting the mixture over a period of time, which may be several weeks or 1-6 weeks, before testing, analyzing, and/or identifying the biological matter source of identified non-human nucleic acids. Methods for RNA and/or DNA identification and nucleic acids (oligonucleotides, polynucleotides) include NGS (next generation sequencing) including robotic machine microprocessor controlled processing of multiple test samples using RT-PCR, LAMP-PCR, and many other PCR methods which utilize a variety of optical multiplexing methods with one or more optical dyes as probes, TaqMan and various selected polynucleotide primers for specific identification of non-human nucleic acids sequences for viruses such as SARS-CoV-2, bacteria, pathogenic life forms, and microorganisms. [00183] Protein Denaturing Agents Used in Methods and Compositions of the Invention [00184] For example, a protein denaturing agent may comprise a high concentration amount of an aqueous chaotropic salt(s) or an organic compound base or organic compound salt such as urea and/or an alkylated urea(s) or a guanidine salt(s) or a derivatized guanidine compound base(s) or/and guanidine salt(s). [00185] The protein denaturing agent(s) may be used to cause an immediate lysis of biological cells, microorganisms, bacteria, virus particles, and any other infectious organisms that may have come from the biological matter in the biological sample. This lysis activity can be used to help sterilize the biological matter in the biological sample without delay after it is obtained from a human, an animal, or another source/location such as a place or surface which a human may use. Denaturing protein enzymes in the biological sample are one means for inactivating the nuclease enzymes (RNases, DNases) which are capable of rapidly degrading nucleic acids in the biological sample. [00186] Agents which can be used to denature proteins include chaotropic agents and disulfide bond reducers. Chaotropic agents include: Urea of 6 – 8 mol/L, guanidine hydrchloride 6 mol/L, Lithium perchlorate 4.5 mol/L, and Sodium dodecyl sulfate __ mol/L. [00187] Disulfide bond reducers include the following chemicals: 2-Mercaptoethanol, Dithiothreitol, TCEP (tris(2-carboxyethyl)phosphine). [00188] Guanidine exists protonated, as guanidine, in solution at physiological pH Guanidinium chloride (also known as guanidine hydrochloride) has chaotropic properties and is used to denature proteins by unfolding them and there is a linear relationship between concentration of guanidine and free energy of unfolding of proteins. In aqueous solutions containing 6 M guanidine hydrochloride, almost all proteins lose their entire secondary structure and become randomly coiled peptide chains. (Guanidine (Wikipedia, 2020). Guanidine thiocyanate is also used for its denaturing effect on various biological samples. [00189] Guanidine exists protonated, as guanidine, in solution at physiological pH. Depicted below are resonance structures for guanidine core structure where R1-R5 are Hydrogen [00190] [00191]
Figure imgf000050_0001
[00192] Guanidines are a group of organic compounds sharing a common functional group with the general structure (R1R2N)(R3R4N)C=N−R5. The guanidine core structure groups R1, R2, R32, R4, and R5 may be selected from the group consisting of hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, 2-pentyl, 3-pentyl, isopentyl, tert-pentyl, straight chain alkyl C6-C10 groups, branched chain alkyl C6-C10 groups, straight chain alkene C1-C10 groups, branched chain alkene C1-C10 groups, C5-C8 cycloalkyl groups, C5-C8 cycloalkenyl groups, C5-C8 cycloaryl groups, polyhexamethylene, and any combination thereof. The central bond within this group is that of an imine, and the group is related structurally to amidines and ureas. [00193] Core Structure of Guanidine and its R1, R2, R3, R4, and R5 Derivatives [00194]
Figure imgf000051_0001
[00195] Chaotropic Anions - For some embodiments of the present invention the guanidine organic cation or another chaotropic cation in a lysis buffer embodiment of the present invention is accompanied by a chaotropic anion. A chaotropic anion may be selected from the group consisting of chloride, nitrate, bromide, iodide, thiocyanate, chlorate, acetate, fluoride, chlorite, thiocyanate, phosphate, hydrogen phosphate, dihydrogen phosphate, propionate, butyrate, nitrite, benzoate, sulfite, sulfide, sulfate, sulfonate, formate, methanoate, glycerate, citrate, malate, malonate, urate, succinate, oxalate, carbonate, bicarbonate, thiophosphate, ferrocyanide, pyrophosphate, and a combination thereof. [00196] For some embodiments of the present invention the guanidine salt for the lysis buffer can be selected from the group consisting of guanidine hydrochloride, guanidine chloride, guanidine nitrate, guanidine bromide, guanidine iodide, guanidine thiocyanate, guanidine chlorate, guanidine acetate, guanidine sulfate, guanidine fluoride, guanidine chlorite, guanidine thiocyanate, guanidine phosphate, guanidine hydrogen phosphate, guanidine dihydrogen phosphate, guanidine propionate, guanidine butyrate, guanidine nitrite, guanidine benzoate, guanidine sulfite, guanidine sulfide, guanidine formate, guanidine methanoate, guanidine glyceride, guanidine citrate, guanidine malate, guanidine malonate, guanidine urate, guanidine succinate, guanidine oxalate, guanidine sulfonate, guanidine carbonate, guanidine bicarbonate, guanidine thiophosphate, guanidine ferrocyanide, guanidine pyrophosphate, guanidine 1-hydroxy-2-naphthoate, guanidine 2,2-dichloroacetate, guanidine 2- hydroxyethanesulfonate, guanidine 2-oxoglutarate, guanidine 4-acetamidobenzoate, guanidine 4-aminosalicylate, guanidine adipate, guanidine ascorbate, guanidine aspartate, guanidine benzenesulfonate, guanidine camphorate, guanidine camphor-10- sulfonate, guanidine decanoate, guanidine hexanoate, guanidine octanoate, guanidine cinnamate, guanidine, guanidine cyclamate, guanidine dodecylsulfurate, guanidine ethane-1,2-disulfonate, guanidine ethanesulfonate, guanidine fumarate, guanidine galactarate, guanidine gentisate, guanidine glucoheptonate, guanidine gluconate, guanidine glucoronate, guanidine glutamate, guanidine glutarate, guanidine glycerophosphoric acid, guanidine glycolate, guanidine hippurate, guanidine isobutyrate, guanidine lactate, guanidine lactobionate, guanidine lauric acid, guanidine mandelate, guanidine methanesulfonate, guanidine naphthalene-1,5-disulfonate, guanidine naphthalene-2-sulfonate, guanidine nicotinate, guanidine oleate, guanidine palmitate, guanidine pamoate, guanidine pyroglutamate, guanidine sebacate, guanidine stearate, guanidine tartarate, guanidine toluenesulfonate, guanidine undecylenate, and a combination thereof. [00197] Typical lysis buffer embodiments of the present invention contain one or more different anions in concentrations between 0.01 molar to 5 molar. Preferably, the lysis buffer contains a mixture of anions wherein each anion is present in an amount less than 4 molar, more preferably wherein each anion is present in an amount less than 2.5 molar. Some lysis buffer embodiments of the present invention have an anion selected from the group consisting of a hydrochloride, a hydrobromide, a hydroiodide, a sulfate, a nitrate, a hydrogen phosphate, a phosphate, an acetate, a propionate, a hexanoate, cyclopentanepropionate, glycolate, a pyruvate, a lactate, malonate, a succinate, maliate, maleate, fumarate, tartrate, a citrate, benzoate, 3-(4-hydroxybenzoyl) benzoate, cinnaminate, mandelate, methanesulfonate, a besylate, an ethanesulfonate, 1,2- ethanedisulfonate, 2-hydroxyethane-sulfonate, benzenesulfonate, 4- chlorobenzenesulfonate, 2-naphthalenesulfonate, 4-toluene-sulfonate, camphorsulfonate, 4-methylbicyclo[2.2.2]-oct-2-ene-1-carboxylate, gluco-heptonate, 3-phenylpropionate, trimethylacetate, tertiary butylacetate, lauryl sulfate, gluconate, glutamate, hydroxynaphthate, salicylate, stearate, muconate, a coordination complex with ethan-olamine, a coordination complex with diethanolamine, a coordination complex with triethanol-amine, a coordination complex with N-methylglucamine, a coordination complex with NTA, a coordination complex with HEPTA, a coordination complex with EGTA, a coordination complex with EDTA, a 2-napsylate, a 3-hydroxy- 2-naphthoate, a 3-phenylprop-ionate, a 4-acetamido-benzoate, an acefyllinate, an aceturate, an adipate, an alginate, an aminosalicylate, an amsonate, an ascorbate, an aspartate, a bicarbonate, a bisulfate, a bi-tartrate, a borate, a butyrate, a calcium edetate, a camphocarbonate, a camphorate, a camsy-late, a carbonate, a cholate, a clavulariate, a cyclopentane-propionate, a cypionate, a d-asp-artate, a d-camsylate, a d-lactate, a decanoate, a dichloroacetate, a digluconate, a dodecyl-sulfate, an edisylate, an estolate, an esylate, an ethyl sulfate, a furate, a fusidate, a galactarate, a mucate, a galacturonate, a gallate, a gentisate, a gluceptate, a glucoheptanoate, a gluconate, a glucuronate, a glutamate, a glutarate, a glycerophosphate, a glycolate, a glycollylarsanilate, a hemisulfate, a heptanoate, an enanthate, a heptanoate, a hexafluorophosphate, a hexanoate, a hexylresorcinate, a hippurate, a hydrabenzate, a hydrabamine, a hydroxybenzoate, a hydroxynaphthoate, a chloride, a bromide, an iodide, a fluoride, an isethionate, an isothionate, an l-aspartate, a l-camsylate, an l- lactate, a lactate, a lactobionate, a laurate, a lauryl-sulphonate, a meso-tartrate, a mesylate, a methanesulfonate, a methylbromide, a methyl-nitrate, a methylsulfate, a myristate, an N-methylglucamine an ammonium salt, a napadisilate, a naphthylate, a napsylate, a nicotinate, an octanoate, an oleate, an orotate, an oxalate, a palmitate, a pamoate, a pantothenate, a pectinate, a persulfate, a phenylpropionate, a phosphateldiphosphate, a picrate, a pivalate, a polygalacturonate, a pyrophosphate, a saccharate, a salicylsulfate, a subacetate, a sulfosaliculate, a sulfosalicylate, a suramate, a tannate, a teoclate, a terephthalate, a thiocyanate, a thiosalicylate, a tosylate, a tribrophenate, a triethiodide, an undecanoate, an undecylenate, a valerate, a valproate, a xinafoate, thioctic acid salt, a folate, and a combination thereof. [00198] Typical lysis buffer embodiments of the present invention contain one or more different cations in concentrations between 0.01 molar to 5 molar. Preferably, the lysis buffer contains a mixture of cations wherein each anion is present in an amount less than 4 molar, more preferably wherein each cation is present in an amount less than 2.5 molar. In some embodiments of the invention the cation has a concentration of less than 0.5 molar. Some lysis buffer embodiments of the present invention have a cation selected from the group consisting of aluminum ion, ammonium ion, antimony ion, arsenic ion, barium ion, beryllium ion, bismuth ion, boron ion, bromide ion, cadmium ion, calcium ion, cerium ion, cesium cation, chloride ion, chromium ion, cobalt ion, copper ion, dysprosium ion, erbium ion, europium ion, fluoride ion, gadolinium ion, gallium ion, germanium ion, gold ion, hafnium ion, holmium ion, indium ion, iodine ion, iridium ion, iron ion, lanthanum ion, lead ion, lithium ion, lutetium ion, magnesium ion, manganese ion, mercury ion, molybdenum ion, neodymium ion, nickel ion, niobium ion, osmium ion, palladium ion, phosphorus ion, platinum ion, potassium ion, praseodymium ion, rhenium ion, rhodium ion, rubidium ion, ruthenium ion, samarium ion, scandium ion, selenium ion, silicon ion, silver ion, sodium ion, strontium ion, sulfate ion, tantalum ion, tellurium ion, terbium ion, thallium ion, thorium ion, thulium ion, tin ion, titanium ion, tungsten ion, vanadium ion, ytterbium ion, yttrium ion, zinc ion, and zirconium ion. Some lysis buffer embodiments comprise at least one inorganic salt with a cation which is not an organic cations having a nitrogen atom, but which is an inorganic cation selected from the group consisting of cesium, lithium, sodium, potassium, iron, zinc, manganese, cobalt, nickel, and copper. [00199] Invention Embodiments of Lysis Buffer Comprise a Protein Denaturing Agent [00200] The lysis buffer may comprise a high concentration of an aqueous chaotropic salt solution for some embodiments of the present invention, wherein the concentration is between about 1 to about 10 molar; about 4 to about 8 molar, or about 4 to about 6 molar. [00201] Example Embodiment of a Lysis Buffer of the present Invention [00202] In one embodiment, the buffer needs to be prepared using water free of ribonucleases (RNases). Examples of RNase-free water may be selected from the group consisting of a medical grade water, a molecular biology grade water, a water treated by reverse osmosis, a double distillation water, an ultrafiltration water, a sterilized water, a membrane filtration water, a charcoal filtered water, a water used for the preparation of injections, a renal dialysis water, an intravenous use water, and any combination thereof. It is conceived as a precaution that the RNase-free water may be purified using an column comprising RNA covalently linked to a solid substrate such as beads as a means for lowering RNase levels in the water to be used to make the buffer. For example, an immobilized RNA-containing column or disk can be attached to a syringe containing a water sample to push through the RNA-containing column so as to remove RNases from the water. [00203] Example composition using an example lysis buffer [00204] The buffer may comprise an RNase-free water and between about 2 M and 6 M guanidine thiocyanate. The buffer may further comprise for example a concentration of sodium citrate between about 1 mM to about 100 mM with a pH 6-7. The buffer may further comprise from 0.1 mM to about 100 mM EDTA to help inhibit DNases if DNA is also being extracted for its identification. [00205] Prepare a silica-based spin column. Silica-based spin columns for the isolation of nucleic acids from biological samples are commercially available for example from: Qiagen, ThermoFisher, Zymo, Macherey Nagel. Prepare a collection tube, also referred to as microcentrifuge tube or Eppendorf tube. Silica-based spin columns and collection tubes are frequently supplied together in a single kit. [00206] Prepare a biological specimen collection swab. Aliquot between about 100 µl and 1000 µl of the lysis buffer, and preferably between about 400 µl and 500 µl, into a sample collection tube. Optionally the sample collection tube is large enough to hold an entire specimen swab. Optionally a specimen swab can be used that comprises a top portion (the handle) that can be broken off so that the swab portion of the specimen swab fits completely into a sample collection tube, thus allowing the sample collection tube to be fully closed with a cap or other type of seal, such as a rubber stop. Perform a specimen collection from the patient or individual being sampled using the specimen swab, such as described previously in the specification. To avoid contamination and risk of infection, do not allow the specimen swab with the collected specimen to come into contact with the outside of the sample collection tube or its cap. [00207] Place the specimen swab into the sample collection tube containing between about 100 µl and 1000 µl, and preferably between about 400 µl and 500 µl, lysis buffer. The lysis buffer, also termed “RELY” buffer “RELY SAFE” buffer, or “RELY” can be used in downstream testing assays in its entirety, thereby allowing for an approximately 4x (four-fold) increase in detection sensitivity compared to protocols where the biological matter is diluted into a fluid, such as VTM, UTM or PBS, in order to prepare a biological sample for testing or analysis. Adding the biological matter specimen directly and immediately into a lysis buffer makes the SARS-CoV-2 nucleic acids assay more sensitive for RNA detection, which reduces false negative results - enables earlier detection. Examples of lower detection limits when using the lysis buffer of the present invention are about 1 – 100 virions per mL. There is no need for refrigeration of a biological sample mixture when the present invention is practiced. An example lysis buffer embodiment of the present invention to prepare the biological sample mixture is depicted in FIG.2C. [00208] Without exception, current methods for detecting DNA and RNA in a biological sample include use of a lysis buffer requiring (1) a very high concentration of a chaotropic salt(s) or urea as a denaturing agent, and (2) a disulfide reducing agent as a lysis composition suitable to inactivate ribonuclease in nucleic acids containing biological matter specimens. Currently available compositions include the disulfide reducing agent, to avoid rapid enzymatic degradation of the nucleic acids by ribonucleases in the biological matter or sample specimen. [00209] Embodiments of the present invention are effective lysis buffer compositions without a reducing agent. A surprising finding of the present invention is that that omitting the beta-mercaptoethanol disulfide reducing agent did not matter! Even without a disulfide reducing agent, the lysis buffer was still fully effective in lysing cells, denaturing proteins and protecting nucleic acids from ribonucleases. The present invention lysis buffer embodiments have the advantage in lacking a disulfide reducing agent because reducing agents such as beta-mercaptoethanol are odorous, volatile, unstable (must be bought fresh frequently) and potentially toxic to human use. Examples of disulfide bond reducers are 2-Mercaptoethanol, Dithiothreitol (DTT), TCEP (tris-2-carboxyethyl phosphine), dithiobutyl-amine (DTBA), and a combination thereof. [00210] However, using a lysis buffer containing guanidine alone or using a disulfide reducing agent is problematic. For example - See Bates US 2011/0027862 A1 which is incorporated herein in its entirety. Paragraph [007] of Bates states: “There are several methods for inhibiting the activity of RNases such as using: (i) ribonuclease peptide inhibitors (“RNasinR”) an expensive reagent only available in small amounts and specific for RNase A, B and C, (ii) reducing agents such as DTT and B- mercaptoethanol which disrupt disulphide bonds in the RNase enzyme, but the effect is limited and temporary as well as being toxic and volatile, (iii) proteases such as proteinase K to digest the RNases, but the transport of proteinases in kits and their generally slow action allows the analyte biomolecules to degrade.” [00211] Bates (2011) also states at paragraph [0011]: “Traditionally RNA degradation is avoided by keeping the contact time between the guanidine and the lysate containing the RNA to a minimum; sample lysis is generally immediately followed by separation of the RNA from the guanidine. Technically this has led to significant problems notably that sample lysis has to be immediately followed by RNA purification which is not always possible or desirable particularly with large numbers of samples, when the assay is a bDNA assay or when automation is involved. It is not always possible to purify RNA at the time or site where the sample is extracted, for example a biopsy from a hospital operating theatre or a blood sample from a doctor’s office. In these cases, the sample must be very carefully stored prior to RNA extraction, which might be carried out within as little as 30 minutes but would more commonly occur only after several hours or days. As a consequence, it has been necessary to develop separate sample storage conditions for each type of tissue and final use of the RNA. As already stated this generally involves using either a dedicated Stabilization Solution, such as RNAlaterTM or PAXgeneTM, or simply immediately freezing the sample in liquid nitrogen.” [00212] PCR methods are continuously being challenged and improved. On July 13, 2020, the CDC published a CDC 2019-Novel Coronavirus (2019-nCoV) Real-Time RT-PCR Diagnostic Panel which is a real-time RT-PCR test intended for the qualitative detection of nucleic acid from the 2019-nCoV in upper and lower respiratory specimens (such as nasopharyngeal or oropharyngeal swabs, sputum, lower respiratory tract aspirates, bronchoalveolar lavage, and nasopharyngeal wash/aspirate or nasal aspirate) collected from individuals who meet 2019-nCoV clinical and/or epidemiological criteria (for example, clinical signs and symptoms associated with 2019-nCoV infection, contact with a probable or confirmed 2019-nCoV case, history of travel to geographic locations where 2019-nCoV cases were detected, or other epidemiologic links for which 2019-nCoV testing may be indicated as part of a public health investigation). The term a biological specimen is the same as the term biological matter. Testing in the United States is limited to laboratories certified under the Clinical Laboratory Improvement Amendments of 1988 (CLIA), 42 U.S.C. § 263a, to perform high complexity tests. Results are for the identification of 2019-nCoV RNA. (See CDC-006-00019, Revision: 05 CDC/DDID/NCIRD/ Division of Viral Diseases Effective: 07/13/2020, Catalog # 2019-nCoVEUA-01, 1000 reactions). [00213] CDC 2019-nCoV Real-Time RT-PCR Diagnostic Panel is a molecular in vitro diagnostic test that aids in the detection and diagnosis 2019-nCoV and is based on widely used nucleic acid amplification technology. The product contains oligonucleotide primers and dual-labeled hydrolysis probes (TaqMan®) and control material used in rRT-PCR for the in vitro qualitative detection of 2019-nCoV RNA in respiratory specimens. The oligonucleotide primers and probes for detection of 2019- nCoV were selected from regions of the virus nucleocapsid (N) gene. The panel is designed for specific detection of the 2019-nCoV (two primer/probe sets). An additional primer/probe set to detect the human RNase P gene (RP) in control samples and clinical specimens is also included in the panel. [00214] The CDC procedure is to isolate RNA and purify RNA from upper and lower respiratory specimens. The RNA is then reverse transcribed to cDNA and subsequently amplified in the Applied Biosystems 7500 Fast Dx Real-Time PCR Instrument with SDS version 1.4 software. In this prior art process, the probe anneals to a specific target sequence located between the forward and reverse primers. During the extension phase of the PCR cycle, the 5’ nuclease activity of Taq polymerase degrades the probe, causing the reporter dye to separate from the quencher dye, generating a fluorescent signal. With each cycle, additional reporter dye molecules are cleaved from their respective probes, increasing the fluorescence intensity. Fluorescence intensity is monitored at each PCR cycle by Applied Biosystems 7500 Fast Dx Real-Time PCR System with SDS version 1.4 software. [00215] Detection of viral RNA not only aids in the diagnosis of illness but also provides epidemiological and surveillance information. [00216] The 2019-nCoV RNA is generally detectable in upper and lower respiratory specimens during infection. Positive results are indicative of active infection with 2019-nCoV but do not rule out bacterial infection or co-infection with other viruses. The agent detected may not be the definite cause of disease. Laboratories within the United States and its territories are required to report all positive results to the appropriate public health authorities. The CDC states that negative results do not preclude 2019-nCoV infection and should not be used as the sole basis for treatment or other patient management decisions. Negative results must be combined with clinical observations, patient history, and epidemiological information. [00217] WHO stability and resistance studies of SARS-CoV-2 virus (SARS coronavirus) based on virus isolation in cell culture and RT-PCR (reverse transcriptase polymerase chain reaction assays) medium and have reported that the SARS-CoV-2 virus is: (1) stable depending on the laboratory for between 6 hours to 2 days in feces and urine at room temperature; (2) stable for 4 days in pH 9 stools from diarrhea patients; (3) has a 10% survival rate after 1 day at room temperature, and after 21 days at 4°C and -80°C in cell-culture supernatant (indicating SARS-CoV-2 virus is more stable than previously-known human coronaviruses); (4) is stable on glass slides for immunofluorescence assays at room temperature unless the acetone used is at -20oC. (https://www.who.int/csr/sars/survival_2003_05_04/en/). [00218] In March, 2020, Relich of CDC established a guidelines mandating human biological sample for SARS-CoV-2 testing shall be placed in a 3 ml screw-capped tube containing a liquid viral transfer medium (VTN) based on their SOP #DSR-052-03 entitled: Preparation of Viral Transport Medium containing 2% fetal bovine serum, Hanks Balanced Salt Solution with calcium and magnesium ions, Gentamycin sulfate, and Amphotericin B and stored at 2-8oC. Detection of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) in clinical samples preserved in phosphate- buffered saline (PBS) or viral transport medium (VTM) was comparable according to Radbel (July, 2020). PBS is a pH 7.2 aqueous solution of only the salts disodium hydrogen phosphate, sodium chloride, potassium chloride, and potassium dihydrogen phosphate (Sigma Aldrich, St. Louis, MO). [00219] Another method for isolating RNA and DNA isas follows: [00220] Step 1. Place the specimen swab into the tube with 450 µl Viral Lysis Buffer. Swirl the swab vigorously for 15 seconds, then expunge as much liquid as possible from the swab by pressing and rotating the fiber portion against the wall of the tube. Discard the swab [00221] Step 2. To each sample, add 10 µl GXP Mag Beads and mix well by pipetting up and down. [00222] Step 3. Incubate the tube for 5 min at 65°C or omit this step if for certain the material at this step contains no active virus. Proteinase K can work better with a small amount of detergent, may not be relevant here [00223] Step 4. Transfer the tube onto the magnetic rack and magnetize the samples for 5 min or until the solution appears clear. [00224] Step 5. Remove and discard the supernatant without disturbing the bead pellet. [00225] Step 6. Add 200 µl Ethanol (80%) to each tube. Do not mix! Incubate for 30 sec. Remove and discard the supernatant without disturbing the bead pellet. [00226] Step 7. Remove and discard the supernatant without disturbing the bead pellet. [00227] Step 8. Add 200 µl Ethanol (80%) to each tube. Do not mix! Incubate for 30 sec. Remove and discard the supernatant without disturbing the bead pellet. Verify the presence of residual ethanol! [00228] Step 9. Remove the tube from the magnet and air dry the pellet for 5-10 min. [00229] Step 10. Add 50 µl Nuclease-free Water to each well and pipetting the sample up and down until the pellet is dissolved. [00230] Step 11. Incubate the tube for 5 min. [00231] Step 12. Transfer the tube onto the magnetic rack and incubate the samples for 3-5 min or until the solution appears clear. [00232] Step 13. Aspirate 50 µl of the supernatant to a new tube. The eluted RNA/DNA can be used immediately for molecular based applications or stored at 80ºC for future use. [00233] Solid Phase Nucleic Acids Extraction on Silica Matrices (Columns / Beads): [00234] Braman (published patent application WO2007149791A2) at paragraph [0149] and its US Patent No.6,180,778 B1 counterpart (both WO2007149791A2 and US6180778 incorporated herein in their entirety) describe various silica matrices that one can use for nucleic acid extraction for the separation of double-stranded/single- stranded nucleic acid structures. [00235] Paragraph [0149] of Braman WO2007149791A2 teaches: “The mineral substrate used for adsorbing a nucleic acid molecule is preferably a filter that comprises or consists of porous or non-porous metal oxides or mixed metal oxides, silica gel, sand, diatomaceous earth, materials predominantly consisting of glass, such as unmodified glass particles, powdered glass, quartz, alumina, zeolites, titanium dioxide, and zirconium dioxide. Fiber filters comprised of glass and any other material that can be molded into a fiber filter may be employed in this method. If alkaline earth metals are used in the mineral substrate, they may be bound by ethylenediaminetetraacetic acid (EDTA) or EGTA, and a sarcosinate may be used as a wetting, washing, or dispersing agent. Any of the materials used for the mineral substrate may also be engineered to have magnetic properties. The particle size of the mineral substrate is preferably from 0.1 um to 1000 um, and the pore size is preferably from 2 to 1000 um. The mineral substrate may be found loose, in filter layers made of glass, quartz, or ceramics, in membranes in which silica gel is arranged, in particles, in fibers, in fabrics of quartz and glass wool, in latex particles, or in frit materials such as polyethylene, polypropylene, and polyvinylidene fluoride especially ultra-high molecular weight polyethylene, high density polyethylene.” [00236] described above are possible without departing from the inventive concept herein. [00237] The heat stable Taq (‘AmpliTaq’) enzyme has a 5’-exonucleolytic function that does not displace the probe sitting in the middle but instead digests it, releasing the reporter dye in the process. Phi29 is a polymerase that is not heat stable (40C max) but which has a strand displacement function. [00238] FIG.14 depicts aspects of the TaqMan kit and process and SYBR Green, and reagents used to detect PCR products. [00239] In another embodiment of the invention the addition of sulfate ions are used to improve the non-infectiousness of the lysis buffer composition and reagent solution for increased safety. In one embodiment of the invention the lysis buffer composition, here called ‘RELY’, provides up to 10x more sensitive testing compared to other collection buffers, such as viral transport medium (VTM), universal transport medium (UTM), physiological or normal saline solution, ‘salina’ buffer solution, phosphate buffered saline (PBS), and others. [00240] Example 5. Sensitivity testing of RELY Lysis Buffer compared to PBS [00241] The control test was done on an AB 7500 qPCR machine using SARS-CoV-2 Viral RNA (Zeptometrix, Cat #0810587CFHI, LOT #324047 with TCID50/ml=3.16E6). 100µl RELY Lysis Buffer or 1000µl PBS (or UTM) or 100µl PBS (or UTM) were used.1µl of 1:100 diluted SARS-CoV-2 (Isolate: USA- WA1/2020) Culture Fluid (Heat Inactivated) from Zeptometrix (Cat #0810587CFHI, LOT #324047) with TCID50/ml=3.16E6) was added as well as 1µl undiluted Plasmid control RNase P (Hs_RPP30) from IDT (Cat #225357026) with 20'000cp/µl in order to simulate a SARS-CoV-2 positive lysate sample as collected from a patient. Direct PCR (without the use of a separate RNA extraction step) was carried out by diluting the lysate sample 1:100 with nuclease-free water. 1µl of the 1:100 diluted lysate sample was used for qPCR and run in triplicate after adding 19µl PCR Mastermix with a Primer/Probe mix for CoV-2 genes N1 and M (RNA) und the human gene RNase P (DNA) as a control. [00242] The volumes used in this control test are considered standard for typical SARS-CoV-2 collection media (4 ml PBS, UTM or VTM), in contrast to a significantly (10x) reduced volume that to be used with RELY (400 µl). To prepare sample for the qPCR reaction, 2µl of each solution of the different lysis buffer compositions were used with 198µl water added (= 1:100 dilution). 2µl of these diluted samples were then run in a total of 20µl qPCR reaction volume. Each difference of 1 in the CT value (cycle threshold qPCR) corresponds to a doubling in concentration. The RELY buffer detects the same viral load at a CT value of 31 compared to a CT of 34.7 in PBS, meaning that the detection sensitivity of RELY is 2^3.7 = 12.6x more sensitive than PBS. [00243] In various embodiments guanidine hydrochloride (GuHCl) was used. In other embodiments guanidine thiocyanate (GuSCN) was used. Other names for GuSCN are guanidine thiocyanate, guanidine thiocyanate or guanidine thiocyanate (GITC). [00244] In one embodiment a solution of 4.5M GuHCl + 0.45M K2SO4 was used. [00245] In another embodiment a solution of 4.5M GuSCN + 0.45M K2SO4 was used. [00246] In another embodiment a solution of 4.5M GuHCl + 0.45M Na2SO4 is used. [00247] In another embodiment a solution of 4.5M GuSCN + 0.45M Na2SO4 is used. [00248] In one embodiment a solution of 4.5M GuHCl + 0.45M K2SO4 is buffered with sodium acetate (NaCH₃COO, also abbreviated NaOAc) to obtain pH 5-6. [00249] In another embodiment a solution of 4.5M GuSCN + 0.45M K2SO4 is buffered with NaOAc to obtain pH 5-6. [00250] In another embodiment a solution of 4.5M GuHCl + 0.45M Na2SO4 is buffered with NaOAc to obtain pH 5-6. [00251] In another embodiment a solution of 4.5M GuSCN + 0.45M Na2SO4 is buffered with NaOAc to obtain pH 5-6. [00252] In one embodiment a solution of 4.5M GuHCl + 0.45M K2SO4 is buffered with Tris (tris(hydroxymethyl)aminomethane, (HOCH2)3CNH2) to obtain pH 7-8.5. [00253] In another embodiment a solution of 4.5M GuSCN + 0.45M K2SO4 is buffered with Tris to obtain pH 7-8.5. [00254] In another embodiment a solution of 4.5M GuHCl + 0.45M Na2SO4 is buffered with Tris to obtain pH 7-8.5. [00255] In another embodiment a solution of 4.5M GuSCN + 0.45M Na2SO4 is buffered with Tris to obtain pH 7-8.5. [00256] In one embodiment a solution of 4.5M GuHCl + 0.45M K2SO4 is buffered with sodium citrate to obtain pH 5-6. ‘Sodium citrate’ can refer to any of the three sodium salts of citric acid. One of these sodium salts of citric acid, trisodium citrate, has the chemical formula Na₃C₆H₅O₇. Another of these sodium salts of citric acid, trisodium citrate, has the chemical formula Na2C6H6O7. A combination of the two can be used to arrive at pH 5-6. Alternatively a solution of HCl (hydrochloric acid) or NaOH (sodium hydroxide) can be used to adjust pH. An example for how to obtain a buffered Tris or TE solution is given below. [00257] In another embodiment a solution of 4.5M GuSCN + 0.45M K2SO4 is buffered with sodium citrate to obtain pH 5-6. [00258] In another embodiment a solution of 4.5M GuHCl + 0.45M Na2SO4 is buffered with sodium citrate to obtain pH 5-6. [00259] In another embodiment a solution of 4.5M GuSCN + 0.45M Na2SO4 is buffered sodium citrate to obtain pH 5-6. Tris Solution 1. Dissolve 121 g Tris base in 800 ml H2O. 2. Adjust to desired pH with concentrated HCl. Approximately 70 ml HCl is needed to achieve a pH 7.4 solution, and 42 ml for a pH 8.0 solution. 3. Adjust volume to 1 liter with H2O. 4. Filter sterilize if necessary. 5. Store up to 6 months at 4°C or room temperature. 1X TE buffer 1. 10 mM Tris, bring to pH 8.0 with HCl. 2. 1 mM EDTA, bring to pH 8.0 with NaOH. [00260] Example 6. FIG.15 shows typical commercially used volumes of viral transport or collection media (4000 µl of UTM, VTM, PBS, salina) were compared to a typical volume of RELY (400 µl). RELY was prepared as 4.5M GuHCl + 0.45M K2SO4 buffered with 1M NaOAc for pH 5 and the N1 viral gene of SARS-CoV-2 was tested in qPCR, comparing the relative sensitivity of RELY vs PBS. Results were run in triplicate and are shown in the table below as “Rep1”, “Rep2”, “Rep3” as well as the Mean value of all three. We get a 3.7 times better CT (cycle threshold qPCR) value for 0.4 ml RELY versus 4 ml PBS. In other words, RELY is 12.6 times more sensitive than other typically used types and volumes of viral collection and transport media and viral lysis buffers. [00261] Gene N1  Rep 1  Rep 2  Rep 3  Mean  RELY  31.6  30.9  30.6  31.0  PBS  34.7  33.9  35.5  34.7  Difference  3.7  Fold  12.6  [00262] In one embodiment, the present RELY buffer has a functional component (sodium sulfate, potassium sulfate, sodium citrate, Tris, NaOAc) that gives it better COVID RNase and human RNase protein insolubility. [00263] In another embodiment, the RELY buffer has an alkaline pH 8-8.5 that gives the SARS-CoV-2 sample less infectivity immediately after collection from a patient. [00264] In another embodiment, the RELY buffer has an acidic pH 5-6 that gives the SARS-CoV-2 sample less infectivity immediately after collection from a patient. [00265] In another embodiment, the RELY buffer contains one ingredient selected from the group of guanidine hydrochloride, guanidine thiocyanate, guanidine sulfate; contains no reducing agent, and contains an additional different function component selected from the group of sodium sulfate, potassium sulfate, sodium citrate, Tris or NaOAc, and any combination thereof. [00266] Preparation of various embodiments of lysate buffer solutions (1-10). The column on the left indicates the type of buffer component that is used with the respective volume in µl given in the right side of the table. For example, a 4.5M GuHCl + 0.45M K2SO4 buffered with 1M NaOAc at pH 5 (column #3, in bold) was generated by adding 90 µl of a stock solution of 5M GuHCl and 0.5M K2SO4, 5 µl of 1 M NaOAc at pH 5, 2.5 µl of SARS-CoV-2 reference (Zeptometrix 1:100), and 2.5 µl RNase P plasmid (IDT 1:1) for a total volume of 100 µl. [00267]
Figure imgf000064_0001
[00268] A data overview of the qPCR results (CT values for the SARS-CoV-2 reference RNA genes ‘N1’ and ‘M’, as well as for the human control DNA gene RNase P (‘RP’) are given below:
Figure imgf000064_0002
0.5M K2SO4 + 
Figure imgf000065_0001
                        A data overview for the SARS-CoV-2 reference RNA gene ‘N1’ is given below:
Figure imgf000065_0002
[00269] FIG.17 is a bar graph providing a data overview of the qPCR results for the SARS-CoV-2 reference RNA gene ‘N1’. [00270] A data overview for the SARS-CoV-2 reference RNA gene ‘M’ is given below:
Figure imgf000065_0003
      [00271] FIG.18 is a bar graph providing a data overview of the qPCR results for the SARS-CoV-2 reference RNA gene ‘M’. [00272] A data overview for the SARS-CoV-2 reference RNA gene ‘M’ is given below:
Figure imgf000065_0004
Figure imgf000066_0001
[00273] FIG.19 is a bar graph providing a data overview of the qPCR results for the human control DNA gene RNase P (‘RP’). [00274] A data overview for the SARS-CoV-2 three reference genes ‘N1’, ‘M’ and ‘RP’ as tested in the RELY formulation No. (see table and description above) is given below: Exp3  Rep 1  Rep 2  Rep 3 
Figure imgf000066_0002
N1  31.6  30.9  30.6  M  35.9  32.7  31.8  RP  36.9  35.8  43.6  [00275] FIG.20 is a bar graph with PCR cycles on left axis needed for COV-2 Genes N1, M1, and RP tested using a RELY Buffer formulation No.3. [00276] A data overview for the SARS-Co-V-2 three reference genes ‘N1’, ‘M’ and ‘RP’ as tested in PBS is given below: Exp10  Rep 1  Rep 2  Rep 3  N1  34.7  33.9  35.5  M  35.6  39.8  41.0  RP  ND  35.5  36.7                      [00277] FIG.21 is a bar graph showing a data overview for the SARS-CoV-2 three reference genes ‘N1’, ‘M’ and ‘RP’ as tested in PBS.     [00278] The qPCR protocol used is given below: Volume per Reaction Buffer Mix (BM) 10 Enzyme Mix (EM) 0.2 Primer / Probe Mix (PPM) 2 Sample or Positiv Control (CTR) 5 Nuclease-free Water 2.8 Total Reaction Volume 20   Step Temp Time Cycle RT 50°C 10min 1 Initial Denaturation 95°C 2min 1 Denaturation 95°C 5sec 50 Anneal/Elongation 60°C 30sec 50 [00279] Additional Lysis Buffer Compositions Embodiments of the Invention: [00280] It is contemplated that the lysis buffer is optimized to solve several potential needs. [00281] Example 7. Lysis Buffer Composition with Cesium cations [00282] In one embodiment of the invention, a lysis buffer consists of an aqueous composition comprising at least 3 ingredients in a volume of a ribonuclease (RNase) free sterile water: [00283] (1) a Cesium salt having a concentration of 0.010 – 4 Molar, of 0.050 – 4 Molar, preferably of 0.1-3 Molar, of 0.1-2 Molar, of 0.1-1 Molar, of 0.1-0.5 Molar; and most preferably of 0.15 to 0.3 Molar; [00284] (2) a pH buffer having a concentration of 0.025-200 millimolar with the pH adjusted to [00285] pH 6-7, to pH 5.9-7.5, and preferably to between pH 6.8-7.1. For example the pH buffer may be sodium citrate, sodium hydrogen phosphate, a Tris Buffer, a “Good” Buffer or another pH buffer. The pH may be adjusted to the desired pH using a base such as sodium hydroxide or potassium hydroxide or any other basic chemical; and [00286] (3) a Guanidine salt having a concentration of between about 5-6 Molar, between about 4-5 Molar, between about 3-4 Molar, between about 2-3 Molar, between about 1-2 Molar, between about 0.5-1 Molar, between about 0.2-0.5 Molar, between about 0.1-0.3 Molar. The guanidine salt is preferably guanidine hydrochloride or guanidine thiocyanate. [00287] (4) Optionally, a multivalent ion chelator such as EDTA, EGTA, NTA, HEDTA and the like may be included having a concentration between about 0.005 to 0.2 Molar. [00288] Example 8. Lysis Buffer Composition with Sodium cations and/or Potassium cations [00289] In one embodiment of the invention, a lysis buffer comprises an aqueous composition comprising at least 3 ingredients in a volume of a ribonuclease (RNase) free sterile water: [00290] (1) a Sodium salt or a potassium salt or a mixture of them having a concentration between about 0.1 – 4 Molar, 0.150 – 3 Molar, between about preferably 0.15 -2 Molar, between about 0.15 -0.1 Molar, between about 0.2-0.5 Molar, between about 0.2-0.3 Molar, more preferably 0.15 to 0.25 Molar. Sodium sulfate and/or potassium sulfate are preferred. [00291] (2) a pH buffer having between about 0.010-.200 Molar pH buffer set to pH 6- 7, between about pH 5.9-7.5, preferably between about pH 6.6-7.2, more preferably between about 6.9-7.1. For example, the pH buffer may be sodium citrate, sodium hydrogen phosphate, a Tris Buffer, a “Good” Buffer or another pH buffer. The pH may be adjusted to the desired pH using a base such as sodium hydroxide or potassium hydroxide; and [00292] (3) a Guanidine salt having a concentration between about 5-6 Molar, between about 4-5 Molar, between about 3-4 Molar, between about 2-3 Molar, between about 1-2 Molar, between about 0.5-1 Molar, between about 0.2-0.5 Molar, between about 0.1-0.3 Molar. The guanidine salt is preferably guanidine hydrochloride or guanidine thiocyanate. [00293] (4) Optionally, a multivalent ion chelator such as EDTA, EGTA, NTA, HEDTA and the like may be includes having a concentration between about 0.005 to 0.2 Molar. [00294] Example 9. Lysis Buffer Composition with Lithium cations [00295] In one embodiment of the invention, a lysis buffer comprises an aqueous composition comprising at least 3 ingredients in a volume of a ribonuclease (RNase) free sterile water: [00296] (1) a Lithium salt having a concentration 0.1 – 4 Molar, 0.150 – 3 Molar, preferably 0.15 -2 Molar, 0.15 -0.1 Molar, 0.2-0.5 Molar, 0.2-0.3 Molar, more preferably 0.15 to 0.25 Molar. Sodium sulfate and/or potassium sulfate are preferred. [00297] (2) a pH buffer having 0.025-200 millimolar of the pH buffer set to pH 6-7, pH 5.9-7.5, preferably to between pH 6.8-7.1. For example the pH buffer may be sodium citrate, sodium hydrogen phosphate, a Tris Buffer, a “Good” Buffer or another pH buffer. The pH may be adjusted to the desired pH using a base such as sodium hydroxide or potassium hydroxide; and [00298] (3) a Guanidine salt having a concentration 5-6 Molar, 4-5 Molar, 3-4 Molar, 2-3 Molar, 1-2 Molar, 0.5-1 Molar, 0.2-0.5 Molar, 0.1-0.3 Molar. The guanidine salt is preferably guanidine hydrochloride or guanidine thiocyanate. [00299] (4) Optionally, a multivalent ion chelators such as EDTA, EGTA, NTA, HEDTA and the like may be includes having a concentration between about 0.005 to 0.2 Molar. [00300] Example 10. Lysis Buffer Composition with Sodium Sulfate [00301] In one embodiment of the invention, a lysis buffer comprises an aqueous composition [00302] comprising at least 3 ingredients in a volume of a ribonuclease (RNase) free sterile water: [00303] (1) Sodium Sulfate salt having a concentration between about 0.05 – 0.2 Molar, 0.150 – 0.3 Molar, preferably 0.15 -0.3 Molar, 0.15 -0.1 Molar, 0.2-0.5 Molar, 0.2-0.3 Molar, more preferably 0.15 to 0.25 Molar. In one embodiment 200 millimolar sodium sulfate salt is used. [00304] (2) a pH buffer having 0.010-200 millimolar of the pH buffer set for example to pH 6.0, 6.2, 6.4, 6.6, 6.8, 7.0, 6-7, pH 5.9-7.5, pH 6.8-7.1. For example the pH buffer may be 25 millimolar sodium citrate buffer, a sodium hydrogen phosphate buffer , a Tris Buffer, a “Good” Buffer or any other pH buffer. The pH may be adjusted using a base such as sodium hydroxide or potassium hydroxide; and [00305] (3) a Guanidine salt having a concentration 5-6 Molar, 4-5 Molar, 3-4 Molar, 2-3 Molar, 1-2 Molar, 0.5-1 Molar, 0.2-0.5 Molar, 0.1-0.3 Molar. In one embodiment the guanidine salt is 5 molar guanidine hydrochloride or 5 molar guanidine thiocyanate. [00306] A Cesium salt may be selected from the group consisting Cesium bromide, Cesium chloride, Cesium iodide, Cesium fluoride, Cesium sulfide, Cesium selenide, Cesium carbonate, Cesium chromate, Cesium dichromate, Cesium dihydrogen phosphate, Cesium bicarbonate, Cesium bisulfate, Cesium monohydrogen phosphate, Cesium nitrate, Cesium nitrite, Cesium perchlorate, Cesium permanganate, Cesium phosphate, Cesium sulfate, Cesium sulfite, Cesium thiosulfate, Cesium silicate, Cesium metasilicate, Cesium aluminum silicate, Cesium acetate, Cesium formate, Cesium oxalate, Cesium methanoate, Cesium glycerate, Cesium citrate, Cesium malate, Cesium malonate, Cesium urate, Cesium succinate, Cesium oxalate, Cesium sulfonate, Cesium thiophosphate, Cesium thiocyanate, Cesium ferrocyanide, Cesium ferricyanide, Cesium pyrophosphate, Cesium gluconate, Cesium benzoate, and a combination thereof. [00307] A Lithium salt may be selected from the group consisting Lithium bromide, Lithium chloride, Lithium iodide, Lithium fluoride, Lithium sulfide, Lithium selenide, Lithium carbonate, Lithium chromate, Lithium dichromate, Lithium dihydrogen phosphate, Lithium bicarbonate, Lithium bisulfate, Lithium monohydrogen phosphate, Lithium nitrate, Lithium nitrite, Lithium perchlorate, Lithium permanganate, Lithium phosphate, Lithium sulfate, Lithium sulfite, Lithium thiosulfate, Lithium silicate, Lithium metasilicate, Lithium aluminum silicate, Lithium acetate, Lithium formate, Lithium oxalate, Lithium methanoate, Lithium glycerate, Lithium citrate, Lithium malate, Lithium malonate, Lithium urate, Lithium succinate, Lithium oxalate, Lithium sulfonate, Lithium thiophosphate, Lithium thiocyanate, Lithium ferrocyanide, Lithium ferricyanide, Lithium pyrophosphate, Lithium benzoate, Lithium gluconate, and a combination thereof. [00308] A Potassium salt may be selected from the group consisting of Potassium bromide, Potassium chloride, Potassium iodide, Potassium fluoride, Potassium sulfide, Potassium selenide, Potassium carbonate, Potassium chromate, Potassium dichromate, Potassium dihydrogen phosphate, Potassium bicarbonate, Potassium bisulfate, Potassium monohydrogen phosphate, Potassium nitrate, Potassium nitrite, Potassium perchlorate, Potassium permanganate, Potassium phosphate, Potassium sulfate, Potassium sulfite, Potassium thiosulfate, Potassium silicate, Potassium metasilicate, Potassium aluminum silicate, Potassium acetate, Potassium formate, Potassium oxalate, Potassium methanoate, Potassium glycerate, Potassium citrate, Potassium malate, Potassium malonate, Potassium urate, Potassium succinate, Potassium oxalate, Potassium sulfonate, Potassium thiophosphate, Potassium thiocyanate, Potassium ferrocyanide, Potassium ferricyanide, Potassium pyrophosphate, Potassium benzoate, Potassium gluconate, and a combination thereof. [00309] A Sodium salt may be selected from the group consisting of Sodium bromide, Sodium chloride, Sodium, Sodium fluoride, Sodium sulfide, Sodium selenide, Sodium carbonate, Sodium chromate, Sodium dichromate, Sodium dihydrogen phosphate, Sodium bicarbonate, Sodium bisulfate, Sodium monohydrogen phosphate, Sodium nitrate, Sodium nitrite, Sodium perchlorate, Sodium permanganate, Sodium phosphate, Sodium sulfate, Sodium sulfite, Sodium thiosulfate, Sodium silicate, Sodium metasilicate, Sodium aluminum silicate, Sodium acetate, Sodium formate, Sodium oxalate, Sodium methanoate, Sodium glycerate, Sodium citrate, Sodium malate, Sodium malonate, Sodium urate, Sodium succinate, Sodium oxalate, Sodium sulfonate, Sodium thiophosphate, Sodium thiocyanate, Sodium ferrocyanide, Sodium ferricyanide, Sodium pyrophosphate, Sodium benzoate, sodium gluconate, and a combination thereof. [00310] Example 11. [00311] The table below shows Storage Stability Testing conducted with the variable of storage temperature on how well RELY Lysis Buffer Compared to PBS Buffer acts to protect COV-2 Zeptometrix gene N1 and COV-2 Zeptrometrix gene M1. Three temperatures were tested: RT(room temperature approximately 20°C), 35°C and 50°C. These temperatures can easily occur in parts of the world without air conditioning such as during storage in a truck.
Figure imgf000071_0001
[00312] Amplification plots are created when the fluorescent signal from each sample is plotted against cycle number; therefore, amplification plots represent the accumulation of product over the duration of the real-time PCR experiment. https://www.qiagen.com/us/resources/faq?id=ee18399a-b88b-43ef-9929- 27d79ef9ed09&lang=en The Rn value, or normalized reporter value, is the fluorescent signal from SYBR Green normalized to (divided by) the signal of the passive reference dye for a given reaction. The delta Rn value is the Rn value of an experimental reaction minus the Rn value of the baseline signal generated by the instrument. This parameter reliably calculates the magnitude of the specific signal generated from a given set of PCR conditions. In our case RT means reverse transcriptase (RT-qPCR = reverse transcriptase quantitative polymerase chain reaction). I. Real-time PCR handbook - Thermo Fisher Scientific II. Essentials of Real-Time PCR | Thermo Fisher Scientific – US www.thermofisher.com › pcr › real-time-pcr-basics › es... [00313] Example 12. [00314] Below is a table showing Storage Stability Testing conducted with the variable of storage temperature on how well RELY Lysis Buffer Compared to PBS Buffer acts to protect COV-2 Zeptometrix gene N1. Three temperatures were tested: RT(room temperature approximately 20°C), 35°C and 50°C. These temperatures can easily occur in parts of the world without air conditioning such as during storage in a truck. [00315] [00316]
Figure imgf000072_0001
[00317] Temperature Resistant Effectiveness of RELY Buffer Means Full Protection RNA at 20 °C, 35 °C, and 50 °C. [00318] As the Temperature is increased in this Experiment with PBS buffers, there was COV gene N1 breakdown in titer due to RNases. This shows the need for there to be more cycles RT-qPCR for a Green SYBR 1 Fluorescence light signal (Rn) to rise off the Rn baseline. [00319] FIG.22 is a graph of storage stability testing conducted with variable storage temperatures on showing how well RELY Lysis Buffer Compared to PBS Buffer acts to protect COV-2 Zeptometrix gene N1. Three temperatures were tested: RT(room temperature approximately 20°C), 35°C and 50°C. [00320] Example 13. [00321] FIG.23 Is a graph of Storage Stability Testing conducted at variable storage temperatures showing how well RELY Lysis Buffer Compared to PBS Buffer acts to protect COV-2 Zeptometrix gene M1. Three temperatures were tested: RT(room temperature approximately 20°C), 35°C and 50°C. These temperatures can easily occur in parts of the world without air conditioning such as during storage in a truck. [00322] The temperature resistant effectiveness of RELY Buffer means full protection of RNA at 20 °C, 35 °C, and 50 °C. [00323] As the Temperature is increased in this Experiment with PBS buffers, there was COV gene M1 breakdown in titer due to RNases. This shows the need for there to be more cycles RT-qPCR for a Green SYBR 1 Fluorescence light signal (Rn) to rise off the Rn baseline. [00324] It will be appreciated by those skilled in the art that changes could be made to the embodiments described above without departing from the broad inventive concept thereof. [00325] It is understood, therefore, that this invention is not limited to the particular embodiments disclosed, but it is intended to cover modifications within the spirit and scope of the present invention as defined by the appended claims. Certain terminology may be used in the following description for convenience only and is not limiting. The words "lower" and "upper" and "top" and "bottom" designate directions in the drawings to which reference is made. The terminology includes the words above specifically mentioned, derivatives thereof and words of similar import. Where a term is provided in the singular, the inventors also contemplate aspects of the invention described by the plural of that term. As used in this specification and in the appended claims, the singular forms "a", "an" and "the" include plural references unless the context clearly dictates otherwise, e.g., "a tip" includes a plurality of tips. Thus, for example, a reference to "a method" includes one or more methods, and/or steps of the type described herein and/or which will become apparent to those persons skilled in the art upon reading this disclosure. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods, constructs and materials are now described. All publications mentioned herein are incorporated herein by reference in their entirety. Where there are discrepancies in terms and definitions used in references that are incorporated by reference, the terms used in this application shall have the definitions given herein.

Claims

What is claimed is: 1. A composition for detecting the presence of viral ribonucleic acid (RNA) in a biological sample comprising a lysis buffer having a protein denaturing agent wherein the protein denaturing agent inactivates proteins in the mixture, prevents degradation of nucleic acids, sterilizes the biological sample without the need for refrigeration, and wherein the lysis buffer does not include a reducing agent.
2. The composition of claim 1, wherein the protein denaturing agent is a chaotropic agent.
3. The composition of claim 2, wherein the chaotropic agent is a chaotropic salt, an organic compound base, or an organic compound salt.
4. The composition of claim 3, wherein the chaotropic salt is a guanidine salt.
5. The composition of claim 2, wherein the chaotropic agent is a derivatized guanidine compound base.
6. The composition of claim 2, wherein the chaotropic agent is selected from the group consisting of urea, guanidine hydrochloride, guanidine thiocyanate, lithium perchlorate, and a combination thereof.
7. The composition of claim 6, wherein the chaotropic agent is selected from the group consisting of urea in a concentration of about 6 mol/L to about 9 mol/L, guanidine hydrochloride in a concentration of about 2.5 mol/L to about 6 mol/L, guanidine thiocyanate in a concentration of about 2.5 mol/L to about 6 mol/L, and lithium perchlorate in a concentration of about 4.5 mol/L to about 6 mol/L.
8. The composition of claim 3, wherein the organic compound base is urea or an alkylated urea.
9. The composition of claim 1, wherein the viral RNA is a SARS-CoV-2 RNA.
10. The composition of claim 1, wherein the lysis buffer comprises a cation, an anion, or a combination thereof in a concentration of about 0.01 m/L to about 6 mol/L.
11. The composition of claim 10, wherein the anion is selected from the group consisting of chloride, nitrate, bromide, iodide, thiocyanate, chlorate, acetate, fluoride, chlorite, phosphate, hydrogen phosphate, dihydrogen phosphate, propionate, butyrate, nitrite, benzoate, sulfite, sulfide, sulfate, sulfonate, formate, methanoate, glycerate, citrate, malate, malonate, urate, succinate, oxalate, carbonate, bicarbonate, thiophosphate, ferrocyanide, pyrophosphate, and a combination thereof.  
12. The composition of claim 10, wherein the cation is selected from the group consisting of ammonium, lithium, cesium, sodium, potassium, and a combination thereof.
13. The composition of claim 1, wherein the lysis buffer further comprises sulfate ions to reduce risk of infectivity of any viral RNA in a biological sample.
14. The composition of claim 1, wherein the composition has a pH of about 5 to about 8 and the lysis buffer comprises (a) RNase-free water; (b) about 2 mol/L to about 6 mol/L guanidine thiocyanate; (c) about 1 mM to about 100 mM of sodium citrate; and (d) one or more sulfate ions.  
15. The composition of claim 14, wherein the composition has a viral RNA quantitative polymerase chain reaction (qPCR) detection sensitivity of about 10x to about 12x more sensitive than solutions selected from the group consisting of viral transport medium (VTM), universal transport medium (UTM), and phosphate buffered saline (PBS).
16. The composition of claim 1, wherein the lysis buffer has an acidic pH of about 5 to about 6 to reduce infectivity of any viral ribonucleic acids in the biological sample.
17. The composition of claim 1, wherein the lysis buffer comprises a guanidine salt; contains no reducing agent; and pH buffer selected from the group consisting of sodium citrate, Tris, NaOAc, and a combination thereof.
18. The composition of claim 1, wherein the lysis buffer includes at least one chaotropic agent and at least one sulfate ion, and the composition is buffered with sodium acetate to obtain a pH of about 5 to about 7.  
19. The composition of claim 1, wherein the lysis buffer includes at least one chaotropic agent and at least one sulfate ion, and the composition is buffered with sodium citrate to obtain a pH of about 5 to about 7.
20. A method for detecting a viral ribonucleic acid (RNA) in a biological sample comprising: (a) obtaining a biological sample from a patient; (b) contacting the biological sample with a lysis buffer including a protein denaturing agent wherein the lysis buffer prevents degradation of nucleic acids, and sterilizes the biological sample, and wherein the lysis buffer does not include a reducing agent and the biological sample is not diluted before contacting the biological sample with the lysis buffer; (c) reverse transcribing the RNA to obtain deoxyribonucleic acid (DNA); and (d) performing a polymerase chain reaction (PCR) to amplify and identify the DNA thereby detecting the presence of the RNA virus nucleic acid in the biological sample.
21. The method of claim 21, wherein the viral RNA is a SARS-CoV-2 RNA.
22. A kit comprising the composition of claim 1.
PCT/US2021/044781 2020-08-05 2021-08-05 Lysis buffer compositions and methods for preparing a viral biological sample useful for covid-19 testing WO2022031992A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202063103435P 2020-08-05 2020-08-05
US63/103,435 2020-08-05
US202063205592P 2020-12-24 2020-12-24
US63/205,592 2020-12-24

Publications (1)

Publication Number Publication Date
WO2022031992A1 true WO2022031992A1 (en) 2022-02-10

Family

ID=80117678

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/044781 WO2022031992A1 (en) 2020-08-05 2021-08-05 Lysis buffer compositions and methods for preparing a viral biological sample useful for covid-19 testing

Country Status (1)

Country Link
WO (1) WO2022031992A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023205353A1 (en) * 2022-04-21 2023-10-26 Ultima Genomics, Inc. Self assembly of beads on substrates

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1081673C (en) * 1996-06-20 2002-03-27 上海市南汇县南华医院 Method and agent box for determining gene hemorrhagic fever virus of nephropathic syndrome
US20090233309A1 (en) * 2007-10-01 2009-09-17 Longhorn Vaccines & Diagnostics, Llc Biological specimen collection/transport compositions and methods
US20130052721A1 (en) * 2010-02-26 2013-02-28 Qiagen Gmbh Method for isolating rna from a rna and dna containing sample
US20130309679A1 (en) * 2012-04-20 2013-11-21 California Institute Of Technology Fluidic devices and systems for sample preparation or autonomous analysis

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1081673C (en) * 1996-06-20 2002-03-27 上海市南汇县南华医院 Method and agent box for determining gene hemorrhagic fever virus of nephropathic syndrome
US20090233309A1 (en) * 2007-10-01 2009-09-17 Longhorn Vaccines & Diagnostics, Llc Biological specimen collection/transport compositions and methods
US20130052721A1 (en) * 2010-02-26 2013-02-28 Qiagen Gmbh Method for isolating rna from a rna and dna containing sample
US20130309679A1 (en) * 2012-04-20 2013-11-21 California Institute Of Technology Fluidic devices and systems for sample preparation or autonomous analysis

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SCALLAN MARTINA F., DEMPSEY CATHERINE, MACSHARRY JOHN, O’CALLAGHAN ISABELLE, O’CONNOR PAULA M., HORGAN CONOR P., DURACK EDEL, COTT: "Validation of a Lysis Buffer Containing 4 M Guanidinium Thiocyanate (GITC)/ Triton X-100 for Extraction of SARS-CoV-2 RNA for COVID-19 Testing: Comparison of Formulated Lysis Buffers Containing 4 to 6 M GITC, Roche External Lysis Buffer and Qiagen RTL Lysis Buffer", BIORXIV, ELI LILLY KINSALE LTD, 13 April 2020 (2020-04-13), XP055899848, Retrieved from the Internet <URL:https://www.biorxiv.org/content/10.1101/2020.04.05.026435v1.full.pdf> [retrieved on 20220310], DOI: 10.1101/2020.04.05.026435 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023205353A1 (en) * 2022-04-21 2023-10-26 Ultima Genomics, Inc. Self assembly of beads on substrates

Similar Documents

Publication Publication Date Title
CN111363729B (en) RNA virus inactivation preservation solution and application thereof
RU2595421C2 (en) Compositions and methods for detection and identification of nucleic acid sequences in biological samples
US8097419B2 (en) Compositions and method for rapid, real-time detection of influenza A virus (H1N1) swine 2009
US9481912B2 (en) Compositions and methods for detecting and identifying nucleic acid sequences in biological samples
ES2660762T3 (en) Methods and compositions for direct chemical lysis
JP2023522054A (en) Methods, apparatus, and kits for preparing nucleic acid samples for storage and analysis
US20140038174A1 (en) Compositions and Methods for Detecting and Identifying Nucleic Acid Sequences in Biological Samples
Parras-Moltó et al. Methods for enrichment and sequencing of oral viral assemblages: saliva, oral mucosa, and dental plaque viromes
WO2022031992A1 (en) Lysis buffer compositions and methods for preparing a viral biological sample useful for covid-19 testing
US11041215B2 (en) PCR ready compositions and methods for detecting and identifying nucleic acid sequences
US11041216B2 (en) Compositions and methods for detecting and quantifying nucleic acid sequences in blood samples
WO2015183811A1 (en) Apparatus and methods for detecting and identifying nucleic acid sequences in biological samples
US20220064709A1 (en) Compositions and Methods for Screening Biological Samples
KR20230171989A (en) Compositions and methods for storing biological samples
JP2024502387A (en) Method for detecting nucleic acids
JP2023521803A (en) Product for detection of viral nucleic acid and method for detection thereof
US20190323068A1 (en) PCR Ready Compositions and Methods for Screening Biological Samples
JP2019528777A (en) PHI6 internal control composition, device, and method
WO2017091809A1 (en) Compositions and methods for detecting and quantifying nucleic acid sequences in blood samples
Pacifico et al. Transcriptomic analyses of phytoplasmas
Esnault et al. Optimisation of rapid untargeted nanopore DNA virus metagenomics using cell cultures and calves experimentally infected with bovine herpes virus-1
Arjmand et al. COVID-19 Diagnosis: A Comprehensive Review of Current Testing Platforms; Part A
TW202330931A (en) Transport medium for microorganism
WO2022167017A1 (en) Liquid composition for molecular diagnostics by pcr
EP4288563A1 (en) Liquid composition for molecular diagnostics by pcr

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 21854662

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21854662

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 21854662

Country of ref document: EP

Kind code of ref document: A1