The following discussion and analysis of our financial condition and results of operations should be read in conjunction with our unaudited financial statements and related notes included in this Quarterly Report on Form 1O-Q and the audited financial statements and notes thereto as of and for the year ended December 31, 2019 and the related Management's Discussion and Analysis of Financial Condition and Results of Operations, both of which are contained in our Annual Report on Form 10-K filed by us with the Securities and Exchange Commission ("SEC"). This Quarterly Report on Form 10-Q may contain "forward-looking statements" within the meaning of Section 27A of the Securities Act of 1933, as amended, or the Securities Act, and Section 21E of the Securities Exchange Act of 1934, as amended, or the Exchange Act. Such forward-looking statements, which represent our intent, belief, or current expectations, involve risks and uncertainties. We use words such as "may," "will," "expect," "anticipate," "estimate," "intend," "plan," "predict," "potential," "believe," "should" and similar expressions to identify forward-looking statements, although not all forward-looking statements contain these identifying words. Such statements may include, but are not limited to, statements concerning projections about our accounting and finances, plans and objectives for the future, future operating and economic performance and other statements regarding future performance. Although we believe the expectations reflected in these forward-looking statements are reasonable, such statements are inherently subject to risk and we can give no assurances that our expectations will prove to be correct. You should not place undue reliance on these forward-looking statements, which apply only as of the date of this Quarterly Report on Form 10-Q. As a result of many factors, including without limitation those set forth under "Risk Factors" under Item 1A of Part II below, and elsewhere in this Quarterly Report on Form 10-Q, our actual results may differ materially from those anticipated in these forward-looking statements. We undertake no obligation to update these forward-looking statements to reflect events or circumstances after the date of this report or to reflect actual outcomes.

References in the following discussion to "we," "our," "us," "Mirati" or "the Company" refer to Mirati Therapeutics, Inc. and its subsidiaries.

Overview

Mirati Therapeutics, Inc. is a clinical-stage oncology company developing product candidates to address the genetic and immunological promoters of cancer. Our KRAS inhibitor programs are focused on developing novel inhibitors of KRAS mutations and includes one clinical program and a preclinical program. In immuno-oncology, we are advancing our kinase inhibitor clinical program where our product candidate has the potential to improve the immune environment of tumor cells and enhance and expand the efficacy of existing cancer immunotherapy medicines when given in combination. We also have additional preclinical programs which include potentially first-in-class and best-in-class product candidates specifically designed to address mutations and tumors where few treatment options exist. We approach each of our discovery and development programs with a singular focus: to translate our deep understanding of the molecular drivers of cancer into better therapies and better outcomes for patients.

Our clinical programs consist of two product candidates: MRTX849, a KRAS G12C inhibitor, and sitravatinib, a multi-kinase inhibitor. We have several early discovery programs, including a preclinical program for a KRAS G12D inhibitor.

KRAS Inhibitor Program

The RAS family of genes is the most commonly mutated oncogene and mutations in this gene family occur in up to approximately 25% of all human cancers. Among the RAS family members, mutations most frequently occur in KRAS (approximately 85% of all RAS family mutations). Tumors characterized by KRAS mutations are commonly associated with poor prognosis and resistance to therapy. Nonclinical studies have demonstrated that cancer cells exhibiting KRAS mutations are highly dependent on KRAS function for cell growth and survival. Historically, KRAS has been extremely difficult to directly inhibit due to the absence of a tractable small molecule drug binding site. Our KRAS inhibitor programs are focused on the discovery and development of small molecule compounds that target KRAS G12C and G12D. We intend to pursue development of our KRAS G12C inhibitor program in both single agent and rational combination approaches. We also have a KRAS G12D inhibitor program in preclinical development.

MRTX849

Background

MRTX849, our lead KRAS G12C compound, is an investigational, specific, potent and orally available small molecule. MRTX849 is designed to directly inhibit KRAS G12C mutations. KRAS G12C mutations are present in approximately 14%



                                       17

--------------------------------------------------------------------------------

Table of Contents

of non-small cell lung cancer ("NSCLC") adenocarcinoma patients, 4% of colorectal cancer ("CRC") patients, 2% of pancreatic cancer patients, as well as smaller percentages of several other difficult-to-treat cancers. Based on observed preclinical attributes, we believe MRTX849 has the potential to be a best-in-class product candidate for the suppression of G12C mutant KRAS signaling. Single agent treatment with MRTX849 has shown complete regression in a subset of KRAS G12C-positive human tumor models implanted in mice.

Program Update

We received U.S. Food and Drug Administration ("FDA") authorization of our investigational new drug application for MRTX849 in November 2018, and on January 15, 2019, we announced that we had dosed the first patient in the dose escalation phase of a Phase 1/2 clinical trial in patients with advanced solid tumors that harbor G12C mutations. This trial is designed to enable expansion of the single agent cohorts and could potentially serve as the basis of a new drug application ("NDA") submission seeking accelerated approval by the FDA. This trial also enables exploratory combination cohorts. Following single agent dose escalation, we are expanding into cohorts that include patients with NSCLC, CRC and those with other tumors that carry the G12C mutation. In the first quarter of 2020, we have initiated enrollment in a registration enabling cohort as a monotherapy in NSCLC.

On October 28, 2019, we reported the first interim clinical data from this Phase 1/2 clinical trial in a presentation at the 2019 American Association for Cancer Research-National Cancer Institute-European Organisation for Research and Treatment of Cancer ("AACR-NCI-EORTC") International Conference on Molecular Targets and Cancer Therapeutics in Boston, Massachusetts. As of October 11, 2019, the trial had enrolled 17 patients, including 10 patients with NSCLC, four patients with CRC, and three patients with other tumor types. Five dose cohorts have been evaluated: 150 mg, 300 mg, 600 mg, and 1200 mg, taken orally once daily ("QD"), and 600 mg, taken orally twice daily ("BID"). The trial enrolled single patient dose escalation cohorts in an accelerated titration design. Trial objectives include evaluation of safety, tolerability, pharmacodynamics, pharmacokinetics ("PK") and tumor response evaluated using RECIST v1.1 criteria.

As of the data cut-off date of October 11, 2019, 12 patients across all dose levels were evaluable for response with at least one radiographic scan.



•      At the highest dose (600 mg BID), three of five evaluable patients with
       NSCLC and one of two evaluable patients with CRC achieved a Partial
       Response ("PR"), and the remaining patients experienced stable disease.



•      Across all dose levels, three of six patients with NSCLC and one of four
       patients with CRC achieved a PR. Two responding patients (one with NSCLC
       and one with CRC) achieved confirmed PRs, both with continuing tumor
       shrinkage following their first scan. The other two patients with PRs
       (both NSCLC) remain on study but have not yet had confirmatory scans.



•      Clinical PK data demonstrated that the dose of 600 mg BID results in drug
       levels that meet or exceed those likely to lead to full inhibition of KRAS
       G12C signaling.



•      Treatment duration across all dose levels ranged from 6.7- 38.6 weeks for
       patients with NSCLC and 9.9-30.1 weeks for patients with CRC as of the
       data cut-off.


Treatment-related adverse events were primarily grade 1 events. One patient experienced a dose-limiting toxicity ("DLT") at the 1200 mg QD dose (capsule burden intolerance 12 capsules) and one patient experienced a DLT at the 600 mg BID dose (grade 3/4 isolated amylase/lipase increase). The maximum tolerated dose was not established and further dose escalation may be explored. Enrollment into dose expansion at the 600 mg BID dose is underway.

MRTX849 Development in Collaboration with Novartis Pharmaceuticals Corporation ("Novartis")

In July 2019, we announced a clinical collaboration agreement with Novartis to evaluate the combination of MRTX849 and Novartis' investigational SHP2 inhibitor, TNO155, in patients with advanced solid tumors that harbor G12C mutations. Under the terms of the non-exclusive collaboration, we will sponsor the trial and we and Novartis will jointly oversee and share the costs of clinical development activities for the combined therapy. Novartis will provide TNO155 at no cost.

MRTX849 and KRAS G12D Discovery Collaboration with Pfizer Inc. ("Pfizer")

In October 2014, we entered into a drug discovery collaboration and option agreement with Array BioPharma, Inc. ("Array," acquired by Pfizer Inc. during 2019) whereby Array provided services to facilitate the discovery, optimization and development of small molecule compounds that bind and specifically inhibit KRAS G12C. In June 2017, the two parties entered into a second, separate discovery collaboration and option agreement whereby Array provided services to facilitate the discovery,



                                       18

--------------------------------------------------------------------------------

Table of Contents

optimization and development of small molecule compounds that bind and specifically inhibit KRAS G12D. Both agreements established an option mechanism which enabled the Company to elect an exclusive worldwide license under the technology for the development and commercialization of certain products based on such compounds.

Under the agreements, following the joint discovery periods which have concluded, we executed our options to retain exclusive worldwide licenses to develop, manufacture and commercialize inhibitors of KRAS G12C and KRAS G12D, including but not limited to, MRTX849 and our KRAS G12D inhibitor program. Under each agreement, Pfizer is entitled to potential development milestone payments of up to $9.3 million, and tiered sales milestone payments of up to $337.0 million based upon worldwide net sales, and tiered royalties in the high single digits to mid-teens on worldwide net sales of products arising from the collaborations. Under the agreements, we have incurred $4.3 million in development milestone payments from inception through March 31, 2020.

Sitravatinib

Sitravatinib is a spectrum-selective kinase inhibitor designed to potently inhibit receptor tyrosine kinases ("RTK"s), including TAM family receptors (TYRO3, Axl, Mer), split family receptors (VEGFR2, KIT) and RET. Sitravatinib is an investigational agent that is being evaluated in combination with immune checkpoint inhibitors.

Sitravatinib's potent inhibition of TAM and split family RTKs may overcome resistance to checkpoint inhibitor therapy through targeted reversal of an immunosuppressive tumor microenvironment, enhancing antigen-specific T cell response and expanding dendritic cell-dependent antigen presentation. As an immuno-oncology agent, sitravatinib is being evaluated in combination with nivolumab (OPDIVO®), Bristol-Myers Squibb Company's ("BMS") anti-PD-1 checkpoint inhibitor, in patients with NSCLC who have experienced documented disease progression following treatment with a checkpoint inhibitor. Sitravatinib is also being developed in certain Asian territories in collaboration with BeiGene, Ltd. ("BeiGene") which is evaluating sitravatinib in combination with tislelizumab, BeiGene's investigational anti-PD-1 checkpoint inhibitor, in a number of advanced solid tumors.

Sitravatinib in Combination with Nivolumab

In an ongoing Phase 2 clinical trial, we are evaluating sitravatinib in combination with nivolumab in patients with NSCLC who have experienced documented disease progression following prior treatment with a checkpoint inhibitor. On May 7, 2020, we announced updated preliminary data from this clinical trial. Patients in the prior clinical benefit ("PCB") cohort experienced PCB on a checkpoint inhibitor as part of their last treatment regimen prior to enrollment. PCB is defined as either complete response, partial response, or stable disease for greater than or equal to 12 weeks. The PCB cohort had been fully enrolled with 87 patients.

As of the data cutoff of January 30, 2020:

• preliminary median overall survival of 15.6 months for the PCB cohort (n=87);





•      preliminary median overall survival of 18.1 months for the subset of PCB
       patients who received the combination as either 2nd or 3rd line of therapy
       after progressing on treatment with a checkpoint inhibitor (n=73), which
       is a patient cohort consistent with the inclusion criteria for the ongoing
       Phase 3 clinical trial; and



•      the combination has been well-tolerated and most adverse events were Grade
       1 or 2 and were similar to data presented previously.


We held an end of Phase 2 meeting with the FDA in the third quarter of 2018 with respect to the development of sitravatinib in combination with a checkpoint inhibitor in NSCLC. Based on feedback received from the FDA, we initiated in July 2019 a Phase 3 randomized clinical trial in second-line NSCLC patients. The Phase 3 clinical trial is comparing the combination of sitravatinib plus nivolumab to docetaxel in patients whose tumors have progressed on prior therapy with platinum-chemotherapy in combination with a checkpoint inhibitor. Ultimately, we expect the results of this clinical trial, if positive, to enable a NDA submission for the treatment of NSCLC patients whose tumors have progressed following treatment with a platinum-containing regimen in combination with a checkpoint inhibitor. Enrollment is ongoing in the Phase 3 clinical trial.

In January 2020, we amended the protocol to include third line patients who have received chemotherapy followed by a checkpoint inhibitor, in addition to second line patients treated with a combination of chemotherapy and a checkpoint inhibitor. We also amended the statistical design to include an interim analysis of overall survival that we believe, if positive, could support an NDA submission seeking full approval. By amending the protocol, the overall sample size decreased from approximately 660 to 530 patients.



                                       19

--------------------------------------------------------------------------------

Table of Contents

On January 7, 2019, we announced a clinical trial collaboration with BMS in connection with the aforementioned Phase 3 clinical trial. Under the terms of the collaboration, we are sponsoring and funding the clinical trial and BMS is providing nivolumab at no cost. In certain specified cases, BMS will have an exclusive right to negotiate a commercial agreement with us for a limited period of time with respect to developing and commercializing sitravatinib worldwide excluding certain territories in Asia, Australia and New Zealand. We maintain global development and commercial rights to sitravatinib outside of certain Asian territories, where we have partnered with BeiGene, and we are free to develop the program in combination with other agents.

During the third quarter of 2018, we initiated an open label, multi-cohort Phase 2 clinical trial of sitravatinib in combination with nivolumab in patients with advanced or metastatic urothelial carcinoma. On November 9, 2019, we reported data from this clinical trial at the 2019 Society of Immunotherapy of Cancer (SITC) 34th Annual Meeting, based on a data cutoff of October 17, 2019. Data from Cohort 1 of the trial were presented, where patients must have been previously treated with an immune checkpoint inhibitor and prior platinum-based chemotherapy and had documented disease progression. A summary of these data is presented below:



•      as of the data cut-off date of October 17, 2019, 22 patients were
       evaluable for response with at least one radiographic scan;



•      6 of 22 evaluable patients achieved a confirmed CR (1 patient) or PR (5
       patients);


• 21 of 22 evaluable patients achieved a confirmed CR, PR, or stable disease;

• 4 responding patients had been treated for more than 6 months; and

• the combination was well-tolerated and most adverse events were Grade 1 or 2.

During the third quarter of 2018, we also initiated an open label Phase 2 clinical trial to assess the mechanism of action of sitravatinib combined with nivolumab in patients with advanced clear cell renal cell cancer ("RCC").

Sitravatinib Development in Collaboration with BeiGene, Ltd.

In January 2018, we entered into a Collaboration and License Agreement (the "BeiGene Agreement") with BeiGene, pursuant to which we and BeiGene agreed to collaboratively develop sitravatinib in Asia (excluding Japan and certain other countries), Australia and New Zealand (the "Licensed Territory"). Under the BeiGene Agreement, we granted BeiGene an exclusive license to develop, manufacture and commercialize sitravatinib in the Licensed Territory, and we retained exclusive rights for the development, manufacturing and commercialization of sitravatinib outside the Licensed Territory.

In November 2018, we announced the dosing of the first patient under the BeiGene Agreement in a Phase 1b clinical trial to assess the safety and tolerability, pharmacokinetics and preliminary anti-tumor activity of sitravatinib in combination with BeiGene's investigational anti-PD-1 antibody, tislelizumab, in patients with advanced solid tumors. The clinical trial is currently enrolling patients in China and Australia. BeiGene's clinical trials will evaluate the combination of sitravatinib and tislelizumab in patients with NSCLC, RCC, hepatocellular cancer, gastric cancer and ovarian cancer. In December 2019, BeiGene reported initial proof of concept data for the ovarian cancer arm of the trial at the 2019 ESMO Immuno-Oncology Congress.

Critical Accounting Policies and Significant Judgments and Estimates Our discussion and analysis of financial condition and results of operations are based upon our consolidated financial statements, which have been prepared in accordance with U.S. generally accepted accounting principles. The preparation of these financial statements requires us to make significant estimates and judgments that affect the reported amounts of assets, liabilities, revenue and expenses and related disclosures. On an ongoing basis, our actual results may differ significantly from our estimates. There have been no material changes to our critical accounting policies and estimates from the information provided in Item 7, Management's Discussion and Analysis of Financial Condition and Results of Operations, included in our Annual Report on Form 10-K for the year ended December 31, 2019.




                                       20

--------------------------------------------------------------------------------

Table of Contents

Results of Operations

Comparison of the Three Months Ended March 31, 2020 and 2019 The following table summarizes the significant items within our results of operations for the three months ended March 31, 2020 and 2019 (in thousands):


                                       Three months ended
                                           March 31,             Increase
                                        2020         2019       (Decrease)

License and collaboration revenues $ 267 $ 1,244 $ (977 )

Research and development expenses $ 71,708 $ 34,240 $ 37,468 General and administrative expenses 18,046 9,762 8,284 Other income, net

                         2,832       1,846            986



Revenues

License and collaboration revenues relate to the BeiGene Agreement under which BeiGene was granted an exclusive license to develop, manufacture and commercialize sitravatinib in the Licensed Territory. License and collaboration revenues for the three months ended March 31, 2020 and 2019 were $0.3 million and $1.2 million, respectively, and relate to revenues earned related to a manufacturing supply services agreement with BeiGene.

Research and development expenses

Research and development expenses consist primarily of:



•            salaries and related expenses for personnel, including expenses
             related to stock options or other share-based compensation granted
             to personnel in research and development functions;


•            fees paid to external service providers such as Clinical Research
             Organizations ("CROs") and contract manufacturing organizations
             related to clinical trials, including contractual obligations for
             clinical development, clinical sites, manufacturing and scale-up,
             and formulation of clinical drug supplies;


•            fees paid to contract services related to drug discovery efforts
             including chemistry and biology services;

• license fees paid in connection with our early discovery efforts; and

• costs for allocated facilities and depreciation of equipment.

We record research and development expenses as incurred.

Our research and development efforts during the three months ended March 31, 2020 and 2019 were focused primarily on our clinical development programs and our preclinical programs. The following table summarizes our research and development expenses, (in thousands):



                                       21

--------------------------------------------------------------------------------

Table of Contents



                                                      Three months ended March 31,       Increase
                                                          2020              2019        (Decrease)
Third-party research and development expenses:
Clinical development programs:
MRTX849                                                      25,090          4,622          20,468
Sitravatinib                                                 15,610         13,729           1,881
Discontinued programs                                           497          1,089            (592 )
Pre-clinical development programs:
KRAS inhibitors                                               5,508          4,438           1,070
Preclinical and early discovery                               3,600            787           2,813

Total third-party research and development expenses 50,305 24,665 25,640 Salaries and other employee related expense

                   7,242          3,816           3,426
Share-based compensation expense                             11,848          5,157           6,691
Other research and development costs                          2,313            602           1,711
Research and development expense                    $        71,708     $   34,240     $    37,468

Research and development expenses for the three months ended March 31, 2020 were $71.7 million compared to $34.2 million for the three months ended March 31, 2019. The increase of $37.5 million primarily relates to increases in third-party research and development expense of $25.6 million, share-based compensation expense of $6.7 million and salaries and other employee related expense of $3.4 million. The increase in third-party research and development expense relates to an increase in expenses associated with development of MRTX849 of $20.5 million and sitravatinib of $1.9 million, offset by decreases in expenses associated with discontinued programs of $0.6 million. The increase in expenses associated with MRTX849 relates to the Phase 1 clinical trial which was initiated in the first quarter of 2019 and the costs are comprised largely of manufacturing expenses, CRO fees and other clinical trial-related expenses. The increase in development expense for sitravatinib is due to increased manufacturing expenses, investigator payment expenses, and CRO expenses to support the expansion of existing and new sitravatinib clinical trials. The decreases in expenses associated with discontinued programs are due to decisions made in prior years to discontinue development of glesatinib and mocetinostat. The increase in share-based compensation of $6.7 million is due to an increase in the fair value of stock options granted during the three months ended March 31, 2020, compared to the same period in 2019. The increase in salaries and other employee related expense of $3.4 million is primarily due to an increase in the number of research and development employees employed during the three months ended March 31, 2020, compared to the same period in 2019.

At this time, due to the risks inherent in the clinical development process and the early stage of our product development programs we are unable to estimate with any certainty the costs we will incur in the continued development of sitravatinib and MRTX849. The process of conducting clinical trials necessary to obtain regulatory approval and manufacturing scale-up to support expanded development and potential future commercialization is costly and time consuming. Any failure by us or delay in completing clinical trials, manufacturing scale up or in obtaining regulatory approvals could lead to increased research and development expense and, in turn, have a material adverse effect on our results of operations. We expect that our research and development expenses may increase if we are successful in advancing sitravatinib, MRTX849 and our preclinical KRAS G12D program, or any of our other preclinical programs into more advanced stages of clinical development.

General and administrative expenses

General and administrative expenses consist primarily of salaries and related benefits, including share-based compensation, related to our executive, finance, business development, legal, human resources and support functions. Other general and administrative expenses include professional fees for auditing, tax, consulting and patent-related services, rent and utilities and insurance.




                                       22

--------------------------------------------------------------------------------

Table of Contents

General and administrative expenses for the three months ended March 31, 2020 and 2019 were $18.0 million and $9.8 million, respectively, representing an increase of $8.2 million. The increase in expense for the three months ended March 31, 2020 is due primarily to an increase in share-based compensation expense of $3.7 million, and to a lesser extent increases in salaries and other employee related expense of $2.7 million, professional services expense of $1.0 million, and facilities, insurance and other expense of $0.8 million. The increase in share-based compensation expense is due to an increase in the fair value of stock options granted during the three months ended March 31, 2020, compared to the same period in 2019. The increase in salaries and other employee related expense and facilities, insurance and other expense is due primarily to an increase in the number of general and administrative employees during the three months ended March 31, 2020 compared to the same period in 2019. The increase in professional services expense is due to an increase in consulting fees.

Other Income, Net

Other income, net for the three months ended March 31, 2020 was $2.8 million compared to $1.8 million for the same period in 2019, and consists primarily of interest income. The increase in interest income is due to an increase in short-term investment balances.



Liquidity and Capital Resources
At March 31, 2020, we had $695.4 million of cash, cash equivalents and
short-term investments compared to $415.1 million at December 31, 2019. In
January 2020, we completed a public offering of our common stock that generated
net proceeds of $324.0 million. Based on our current and anticipated level of
operations, we believe that our cash, cash equivalents and short-term
investments will be sufficient to meet our anticipated obligations for at least
one year from the date that this Quarterly Report on Form 10-Q is filed with the
SEC.
To date, we have funded our operations primarily through the sale of our common
stock, pre-funded warrants to purchase our common stock, and to a lesser extent
through up-front payments, research funding and milestone payments under
collaborative arrangements. Since inception, we have primarily devoted our
resources to funding research and development programs, including discovery
research, preclinical and clinical development activities. To fund future
operations, we will likely need to raise additional capital. The amount and
timing of future funding requirements will depend on many factors, including the
timing and results of our ongoing development efforts, the potential expansion
of our current development programs, potential new development programs and
related general and administrative support. We anticipate that we will seek to
fund our operations through public or private equity or debt financings or other
sources, such as potential collaboration agreements. We cannot make assurances
that anticipated additional financing will be available to us on favorable
terms, or at all. Although we have previously been successful in obtaining
financing through our equity securities offerings, there can be no assurance
that we will be able to do so in the future. As a result of the COVID-19
pandemic and actions taken to slow its spread, the global credit and financial
markets have experienced extreme volatility, including diminished liquidity and
credit availability, declines in consumer confidence, declines in economic
growth, increases in unemployment rates and uncertainty about economic
stability. There can be no assurance that further deterioration in credit and
financial markets and confidence in economic conditions will not occur. If
equity and credit markets deteriorate, it may make any necessary debt or equity
financing more difficult to obtain, more costly and/or more dilutive.

Cash Flows for the Three Months Ended March 31, 2020 and 2019

The following table provides a summary of the net cash flow activity for each of the periods set forth below (in thousands):


                                                          Three months ended
                                                               March 31,
                                                          2020          2019
Net cash used in operating activities                  $ (59,004 )   $ (34,482 )
Net cash used in investing activities                   (122,227 )     (35,208 )
Net cash provided by financing activities                339,201       111,601

Increase in cash, cash equivalents and restricted cash 157,970 41,911

Net cash used in operating activities

Net cash used in operating activities for the three months ended March 31, 2020 was $59.0 million, compared to $34.5 million for the three months ended March 31, 2019, an increase of $24.5 million. Cash used in operating activities during 2020 primarily related to our net loss of $86.7 million, adjusted for non-cash items such as share-based compensation of $21.6 million and net cash inflows from a change in our operating assets and liabilities of $6.5 million. Cash used in operating



                                       23

--------------------------------------------------------------------------------

Table of Contents

activities during 2019 primarily related to our net loss of $40.9 million, adjusted for non-cash items such as share-based compensation expense of $11.1 million and net cash outflows from a change in our operating assets and liabilities of $3.8 million.

Net cash used in investing activities

For the three months ended March 31, 2020 and March 31, 2019, investing activities used cash of $122.2 million and $35.2 million, respectively due to purchases of short-term investments and property and equipment, offset by sales and maturities of short-term investments.

Net cash provided by financing activities

Net cash provided by financing activities for the three months ended March 31, 2020 and 2019 was $339.2 million and $111.6 million, respectively, and consisted of proceeds received from the issuance of common stock, exercise of common stock options, and disgorgement of shareholders' short-swing profits.

Off-Balance Sheet Arrangements

During the three months ended March 31, 2020, we did not have any off-balance sheet arrangements (as defined by applicable SEC regulations) that are reasonably likely to have a current or future material effect on our financial condition, results of operations, liquidity, capital expenditures or capital resources.

Contractual Obligations and Commitments

There were no material changes outside of the ordinary course of business to our specific contractual obligations during the three months ended March 31, 2020.

Recent Accounting Pronouncements

Occasionally, new accounting standards are issued or proposed by the Financial Accounting Standards Board, or other standard-setting bodies that we adopt by the effective date specified within the standard. Unless otherwise discussed, standards that do not require adoption until a future date are not expected to have a material impact on our financial statements upon adoption.

© Edgar Online, source Glimpses